Signal-sensor polynucleotides for the alteration of cellular phenotypes (2024)

This application is a continuation of U.S. patent application Ser. No. 16/036,170 filed Jul. 16, 2018, which is a continuation of U.S. patent application Ser. No. 14/041,011 filed on Sep. 30, 2013, abandoned; which claims priority to U.S. Provisional Patent Application Ser. No. 61/753,661 filed Jan. 17, 2013; U.S. Provisional Patent Application Ser. No. 61/754,159 filed Jan. 18, 2013; U.S. Provisional Patent Application Ser. No. 61/781,097 filed Mar. 14, 2013; U.S. Provisional Patent Application Ser. No. 61/829,334 filed May 31, 2013; U.S. Provisional Patent Application Ser. No. 61/839,893 filed Jun. 27, 2013; U.S. Provisional Patent Application Ser. No. 61/842,733 filed Jul. 3, 2013; and U.S. Provisional Patent Application Ser. No. 61/857,304 filed Jul. 23, 2013. The contents of the aforementioned applications are hereby incorporated by reference in their entireties.

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jan. 29, 2021, is named MDN_037 CN4_SL.txt and is 9748552 bytes in size.

The invention relates to compositions, methods, processes, kits and devices for the design, preparation, manufacture and/or formulation of signal-sensor polynucleotides, primary constructs and mRNA molecules for the alteration of cellular phenotypes and microenvironments.

Cancer is a disease characterized by uncontrolled cell division and growth within the body. In the United States, roughly a third of all women and half of all men will experience cancer in their lifetime. Polypeptides are involved in every aspect of the disease including cancer cell biology (carcinogenesis, cell cycle suppression, DNA repair and angiogenesis), treatment (immunotherapy, hormone manipulation, enzymatic inhibition), diagnosis and determination of cancer type (molecular markers for breast, prostate, colon and cervical cancer for example). With the host of undesired consequences brought about by standard treatments such as chemotherapy and radiotherapy used today, genetic therapy for the manipulation of disease-related peptides and their functions provides a more targeted approach to disease diagnosis, treatment and management.

To this end, it has been previously shown that certain modified mRNA sequences have the potential as therapeutics with benefits beyond just evading, avoiding or diminishing the immune response. Such studies are detailed in published co-pending International Publication No WO2012019168 filed Aug. 5, 201, International Publication No WO2012045082 filed Oct. 3, 2011, International Publication No WO2012045075 filed Oct. 3, 2011, International Publication No WO2013052523 filed Oct. 3, 2012, and International Publication No WO2013090648 filed Dec. 14, 2012 the contents of which are incorporated herein by reference in their entirety.

The use of modified polynucleotides in the fields of antibodies, viruses, veterinary applications and a variety of in vivo settings have been explored and are disclosed in, for example, co-pending and co-owned U.S. Provisional Patent Application No. 61/618,862, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Biologics; U.S. Provisional Patent Application No. 61/681,645, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Biologics; U.S. Provisional Patent Application No. 61/737,130, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Biologics; U.S. Provisional Patent Application No. 61/618,866, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Antibodies; U.S. Provisional Patent Application No. 61/681,647, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Antibodies; U.S. Provisional Patent Application No. 61/737,134, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Antibodies; U.S. Provisional Patent Application No. 61/618,868, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Vaccines; U.S. Provisional Patent Application No. 61/681,648, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Vaccines; U.S. Provisional Patent Application No. 61/737,135, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Vaccines; U.S. Provisional Patent Application No. 61/618,870, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Therapeutic Proteins and Peptides; U.S. Provisional Patent Application No. 61/681,649, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Therapeutic Proteins and Peptides; U.S. Provisional Patent Application No. 61/737,139, filed Dec. 14, 2012, Modified Polynucleotides for the Production of Therapeutic Proteins and Peptides; U.S. Provisional Patent Application No. 61/618,873, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Secreted Proteins; U.S. Provisional Patent Application No. 61/681,650, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Secreted Proteins; U.S. Provisional Patent Application No. 61/737,147, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Secreted Proteins; U.S. Provisional Patent Application No. 61/618,878, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Plasma Membrane Proteins; U.S. Provisional Patent Application No. 61/681,654, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Plasma Membrane Proteins; U.S. Provisional Patent Application No. 61/737,152, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Plasma Membrane Proteins; U.S. Provisional Patent Application No. 61/618,885, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Cytoplasmic and Cytoskeletal Proteins; U.S. Provisional Patent Application No. 61/681,658, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Cytoplasmic and Cytoskeletal Proteins; U.S. Provisional Patent Application No. 61/737,155, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Cytoplasmic and Cytoskeletal Proteins; U.S. Provisional Patent Application No. 61/618,896, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Intracellular Membrane Bound Proteins; U.S. Provisional Patent Application No. 61/668,157, filed Jul. 5, 2012, entitled Modified Polynucleotides for the Production of Intracellular Membrane Bound Proteins; U.S. Provisional Patent Application No. 61/681,661, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Intracellular Membrane Bound Proteins; U.S. Provisional Patent Application No. 61/737,160, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Intracellular Membrane Bound Proteins; U.S. Provisional Patent Application No. 61/618,911, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Nuclear Proteins; U.S. Provisional Patent Application No. 61/681,667, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Nuclear Proteins; U.S. Provisional Patent Application No. 61/737,168, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Nuclear Proteins; U.S. Provisional Patent Application No. 61/618,922, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Proteins; U.S. Provisional Patent Application No. 61/681,675, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Proteins; U.S. Provisional Patent Application No. 61/737,174, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Proteins; U.S. Provisional Patent Application No. 61/618,935, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/681,687, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/737,184, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/618,945, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/681,696, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/737,191, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/618,953, filed Apr. 2, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/681,704, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/737,203, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. Provisional Patent Application No. 61/681,720, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Cosmetic Proteins and Peptides; U.S. Provisional Patent Application No. 61/737,213, filed Dec. 14, 2012, entitled Modified Polynucleotides for the Production of Cosmetic Proteins and Peptides; U.S. Provisional Patent Application No. 61/681,742, filed Aug. 10, 2012, entitled Modified Polynucleotides for the Production of Oncology-Related Proteins and Peptides; International Application No PCT/US2013/030062, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Biologics and Proteins Associated with Human Disease; U.S. patent application Ser. No. 13/791,922, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Biologics and Proteins Associated with Human Disease; International Application No PCT/US2013/030063, filed Mar. 9, 2013, entitled Modified Polynucleotides; International Application No. PCT/US2013/030064, entitled Modified Polynucleotides for the Production of Secreted Proteins; U.S. patent application Ser. No. 13/791,921, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Secreted Proteins; International Application No PCT/US2013/030059, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Membrane Proteins; International Application No. PCT/US2013/030066, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Cytoplasmic and Cytoskeletal Proteins; International Application No. PCT/US2013/030067, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Nuclear Proteins; International Application No. PCT/US2013/030060, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Proteins; International Application No. PCT/US2013/030061, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; U.S. patent application Ser. No. 13/791,910, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Proteins Associated with Human Disease; International Application No. PCT/US2013/030068, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Cosmetic Proteins and Peptides; and International Application No. PCT/US2013/030070, filed Mar. 9, 2013, entitled Modified Polynucleotides for the Production of Oncology-Related Proteins and Peptides; International Patent Application No. PCT/US2013/031821, filed Mar. 15, 2013, entitled In Vivo Production of Proteins; the contents of each of which are herein incorporated by reference in their entireties.

Formulations and delivery of modified polynucleotides are described in, for example, co-pending and co-owned International Publication No WO2013090648, filed Dec. 14, 2012, entitled Modified Nucleoside, Nucleotide, Nucleic Acid Compositions and US Publication No US20130156849, filed Dec. 14, 2012, entitled Modified Nucleoside, Nucleotide, Nucleic Acid Compositions; the contents of each of which are herein incorporated by reference in their entireties.

The next generation of therapeutics must also address the complex cellular microenvironment of the cancer and have the capacity for cell, tissue, organ or patient stratification, whether structurally or functionally.

The present invention addresses this need by providing nucleic acid based compounds or polynucleotide-encoding nucleic acid-based compounds (e.g., signal-sensor polynucleotides) which encode a polypeptide of interest and which have structural and/or chemical features that allow for greater selectivity, profiling or stratification along definable disease characteristics or metrics.

Described herein are compositions, methods, processes, kits and devices for the design, preparation, manufacture and/or formulation of signal-sensor polynucleotide molecules encoding at least one oncology-related polypeptide of interest. Such signal-sensor polynucleotides may be chemically modified mRNA (mmRNA) molecules.

The present invention provides an isolated signal-sensor polynucleotide comprising a region encoding an oncology-related polypeptide of interest that functions, when translated, to send a death or survival signal. Such death or survival signals include those which (i) alter (increase or decrease) the expression of one or more proteins, nucleic acids, or non-coding nucleic acids, (ii) alter the binding properties of biomolecules within the cell, and/or (iii) perturb the cellular microenvironment in a therapeutically beneficial way.

Optionally, the signal-sensor polynucleotide may also encode in a flanking region, one or more sensor sequences. Such sensor sequences function to “sense” the cell, tissue or organ microenvironment and confer upon the signal-sensor polynucleotide an altered expression or half life profile (increased or decreased) depending on the interactions of the sensor sequence with the cell, tissue or organ microenvironment.

In one aspect, provided herein are signal-sensor polynucleotide comprising, a first region of linked nucleosides, a first flanking region located 5′ relative to said first region and a second flanking region located 3′ relative to said first region. The first region may encode an oncology-related polypeptide of interest such as, but not limited to, SEQ ID NOs: 1321-2487, 6611-6616 and 7355-7361, 7490, 7492, 7493, 7512, 7514, 7516 and 7517 and the first flanking region may include a sequence of linked nucleosides such as, but not limited to, the native 5′ untranslated region (UTR) of any of the nucleic acids that encode any of SEQ ID NOs: 1321-2487, 6611-6616, 7355-7361, 7490, 7492, 7493, 7512, 7514, 7516, 7517, SEQ ID NO: 1-4 and functional variants thereof. The first region may comprise at least an open reading frame of a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 2488-2496, 6617-6621, 7348-7354, 7362-7489, 7491, 7494, 7506, 7511 and 7513.

The second flanking region may include a sequence of linked nucleosides such as, but not limited to, the native 3′ UTR of any of the nucleic acids that encode any of SEQ ID NOs: 1321-2487, 6611-6616, 7355-7361, 7490, 7492, 7493, 7512, 7514, 7516, 7517, SEQ ID NO: 5-21 and functional variants thereof, and one or more sensor sequences located such as, but not limited to, SEQ ID NOs: 3529-4549, SEQ ID NOs: 5571-6591 and functional variants thereof. The signal-sensor polynucleotide may also include a 3′ tailing sequence of linked nucleosides.

In another aspect, provided herein is a signal-sensor polynucleotide which comprises an mRNA encoding an oncology-related polypeptide of interest and one or more sensor sequences such as, but not limited to, SEQ ID NOs: 3529-4549, SEQ ID NOs: 5571-6591 and functional variants thereof. The oncology-related polypeptide of interest may be, but is not limited to, SEQ ID NOs: 1321-2487, 6611-6616, 7355-7361, 7490, 7492, 7493, 7512, 7514, 7516 and 7517. The mRNA may include at least one open reading frame of a nucleic acid sequence selected from the group consisting of SEQ ID NOs: 2488-2496, 6617-6621, 7348-7354, 7362-7489, 7491, 7494, 7506, 7511 and 7513.

The signal-sensor polynucleotides may comprise one, two, three or more than three stop codons. In one aspect, the signal-sensor polynucleotides comprise two stop codons. As a non-limiting example, the first stop codon is “TGA” and the second stop codon is selected from the group consisting of “TAA,” “TGA” and “TAG.” In another aspect, signal-sensor polynucleotides comprise three stop codons.

The signal-sensor polynucleotides may have a 3′ tailing sequence of linked nucleosides such as, but not limited to, a poly-A tail of at least 140 nucleotides, a triple helix, and a poly A-G quartet.

The signal-sensor polynucleotides may have a 5′ cap such as, but not limited to, Cap0, Cap1, ARCA, inosine, N1-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.

In one aspect, the signal-sensor polynucleotides may include at least one chemical modification such as, but not limited to, modifications located on one or more of a nucleoside and/or the backbone of the nucleotides. In one embodiment, the signal-sensor polynucleotides comprise a pseudouridine analog such as, but not limited to, 1-carboxymethyl-pseudouridine, 1-propynyl-pseudouridine, 1-taurinomethyl-pseudouridine, 1-taurinomethyl-4-thio-pseudouridine, 1-methyl-pseudouridine (m1ψ), 1-methyl-4-thio-pseudouridine (m1s4ψ), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m3ψ), 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydropseudouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine, 1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine (acp3 ψ), and 2′-O-methyl-pseudouridine (ψm). In another embodiment, the signal-sensor polynucleotides comprise the pseudouridine analog 1-methylpseudouridine. In yet another embodiment, the signal-sensor polynucleotides comprise the pseudouridine analog 1-methylpseudouridine and the modified nucleoside 5-methylcytidine.

In another aspect, the signal-sensor polynucleotides may include at least two chemical modifications such as, but not limited to, modifications located on one or more of a nucleoside and/or the backbone of the nucleotides. As a non-limiting example, the signal-sensor polynucleotide comprises the chemical modifications 1-methylpseudouridine and 5-methylcytidine.

The signal-sensor polynucleotides may comprise at least one translation enhancer element (TEE) such as, but not limited to, TEE-001-TEE-705.

In one aspect, the signal-sensor polynucleotide encodes a factor modulating the affinity between HIF subunits and/or HIF-dependent gene expression such as, but not limited to, SEQ ID NO: 6611-6616.

The signal-sensor polynucleotides may be purified and/or formulated.

Employing the signal-sensor polynucleotides, the present invention provides a method of treating a disease, disorder and/or condition in a subject in need thereof by increasing the level of an oncology-related polypeptide of interest comprising administering to said subject an isolated signal-sensor polynucleotide encoding said oncology-related polypeptide. The disease, disorder and/or condition may include, but is not limited to, adrenal cortical cancer, advanced cancer, anal cancer, aplastic anemia, bileduct cancer, bladder cancer, bone cancer, bone metastasis, brain tumors, brain cancer, breast cancer, childhood cancer, cancer of unknown primary origin, Castleman disease, cervical cancer, colon/rectal cancer, endometrial cancer, esophagus cancer, Ewing family of tumors, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gestational trophoblastic disease, Hodgkin disease, Kaposi sarcoma, renal cell carcinoma, laryngeal and hypopharyngeal cancer, acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, chronic myelomonocytic leukemia, liver cancer, non-small cell lung cancer, small cell lung cancer, lung carcinoid tumor, lymphoma of the skin, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma in adult soft tissue, basal and squamous cell skin cancer, melanoma, small intestine cancer, stomach cancer, testicular cancer, throat cancer, thymus cancer, thyroid cancer, uterine sarcoma, vagin*l cancer, vulvar cancer, Waldenstrom macroglobulinemia, Wilms tumor and secondary cancers caused by cancer treatment.

The present invention provides a method of reducing, eliminating, or preventing tumor growth in a subject in need thereof by increasing the level of an oncology-related polypeptide of interest comprising administering to said subject an isolated signal-sensor polynucleotide encoding said oncology-related polypeptide. The tumor growth may be associated with or results from a disease, disorder and/or condition such as, but not limited to, adrenal cortical cancer, advanced cancer, anal cancer, aplastic anemia, bileduct cancer, bladder cancer, bone cancer, bone metastasis, brain tumors, brain cancer, breast cancer, childhood cancer, cancer of unknown primary origin, Castleman disease, cervical cancer, colon/rectal cancer, endometrial cancer, esophagus cancer, Ewing family of tumors, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gestational trophoblastic disease, Hodgkin disease, Kaposi sarcoma, renal cell carcinoma, laryngeal and hypopharyngeal cancer, acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, chronic myelomonocytic leukemia, liver cancer, non-small cell lung cancer, small cell lung cancer, lung carcinoid tumor, lymphoma of the skin, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma in adult soft tissue, basal and squamous cell skin cancer, melanoma, small intestine cancer, stomach cancer, testicular cancer, throat cancer, thymus cancer, thyroid cancer, uterine sarcoma, vagin*l cancer, vulvar cancer, Waldenstrom macroglobulinemia, Wilms tumor and secondary cancers caused by cancer treatment.

The present invention provides a method of reducing and/or ameliorating at least one symptom of cancer in a subject in need thereof by increasing the level of a polypeptide of interest comprising administering to said subject an isolated signal-sensor polynucleotide encoding said oncology-related polypeptide. Non-limiting examples of symptoms include weakness, aches and pains, fever, fatigue, weight loss, blood clots, increased blood calcium levels, low white blood cell count, short of breath, dizziness, headaches, hyperpigmentation, jaundice, erythema, pruritis, excessive hair growth, change in bowel habits, change in bladder function, long-lasting sores, white patches inside the mouth, white spots on the tongue, unusual bleeding or discharge, thickening or lump on parts of the body, indigestion, trouble swallowing, changes in warts or moles, change in new skin and nagging cough and hoarseness.

The present invention provides a method of preferentially inducing cell death in cancer cells in a tissue or organ comprising contacting the tissue or organ with a signal-sensor polynucleotide encoding an oncology-related polypeptide whose expression triggers apoptosis or cell death and at least one microRNA binding site of a microRNA where the expression of the microRNA in the cancer cell is lower than the expression of the mircroRNA in normal non-cancerous cells.

The signal-sensor polynucleotide may be administered at a total daily dose of between 0.001 ug and 150 ug. Administration of a signal-sensor polynucleotide may be by injection, topical administration, ophthalmic administration or intranasal administration. In one aspect, administration may be by injection such as, but not limited to, intradermal, subcutaneous and intramuscular. In another aspect, administration may be topical such as, but not limited to, using creams, lotions, ointments, gels, sprays, solutions and the like.

The details of various embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and the drawings, and from the claims.

The foregoing and other objects, features and advantages will be apparent from the following description of particular embodiments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of various embodiments of the invention.

FIG. 1 is a schematic of a primary construct of the present invention.

FIG. 2 is an expanded schematic of the second flanking region of a primary construct of the present invention illustrating the signal-sensor elements of the polynucleotide.

FIGS. 3A and 3B are gel profiles of Apoptosis-Inducing Factor short (AIFsh) protein from AIFsh modified mRNA in mammals. FIGS. 3A and 3B show the expected size of AIFsh.

FIGS. 4A and 4B are gel profiles of Siah E3 ubiquitin protein ligase 1 (SIAH1) protein from SIAH1 modified mRNA in mammals. FIGS. 4A and 4B show the expected size of SIAH1.

FIGS. 5A and 5B are gel profiles of constitutively active (C.A.) caspase 3 (also known as reverse caspase 3 (Rev-Caspase 3)) protein from C.A. caspase 3 modified mRNA in mammals. FIGS. 5A and 5B show the expected size of C.A. caspase 3.

FIGS. 6A and 6B are gel profiles of Granulysin protein from granulysin modified mRNA in mammals. FIGS. 6A and 6B show the expected size of granulysin.

FIGS. 7A and 7B are western blots of C.A. caspase 3 and C.A. caspase 6. FIG. 7A shows protein from C.A. caspase 3 modified mRNA fully modified with 5-methylcytidine and 1-methylpseudouridine or fully modified with 1-methylpseudouridine. FIG. 7B shows protein from C.A. caspase 6 modified mRNA fully modified with 5-methylcytidine and 1-methylpseudouridine or fully modified with 1-methylpseudouridine.

It is of great interest in the fields of therapeutics, diagnostics, reagents and for biological assays to be able to deliver a nucleic acid, e.g., a ribonucleic acid (RNA) inside a cell, whether in vitro, in vivo, in situ or ex vivo, such as to cause intracellular translation of the nucleic acid and production of an encoded polypeptide of interest. Of particular importance is the delivery and function of a non-integrative polynucleotide.

Described herein are compositions (including pharmaceutical compositions) and methods for the design, preparation, manufacture and/or formulation of polynucleotides encoding one or more polypeptides of interest. Also provided are systems, processes, devices and kits for the selection, design and/or utilization of the polynucleotides encoding the polypeptides of interest described herein.

To this end, polypeptides of the present invention are encoded by a new class of polynucleotide therapeutics, termed “signal-sensor polynucleotides” which are particularly useful in the stratification, profiling and/or personalization of the polynucleotide therapeutic (e.g., mRNA) and which are tailored to a particular cell type, disease or cell microenvironment or biological profile.

It is known that cancers exhibit diverse gene expression patterns, physicochemical environments and metastatic or motility behaviors and according to Hanahan and Weinberg (Cell, 2011, 144:646-674) there are six hallmarks of cancer. These include sustaining a proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. These hallmarks or functions of cancer allow the cancer to survive, proliferate and disseminate and each arises at different times and in different patterns depending on the cancer type.

The development of cancer therapeutics which to selectively target the cancer cells while sparing normal cells dominates ongoing efforts in every area of oncology. The polynucleotides of the present invention represent such therapeutics; having the ability to selectively stabilize or destabilize cell systems, signal proliferation (survival) or death, trigger the cell cycle or senescence and/or activate or avoid the immune response depending on the cell type, e.g., cancer or normal cell.

According to the present invention, signal-sensor polynucleotide therapeutics may be used to destabilize the survival advantages or hallmarks of a cancer cell (hence they would be cytotoxic). In one embodiment diagnostic efforts would include the profiling of the cancer (although this would not be required a priori) including metabolic state (hypoxic, acidotic), apoptotic vs. survival gene profiles, cell cycle vs. senescent stage, immune status, and stromal factors present.

In one embodiment the signal-sensor polynucleotide disrupts the transcriptome of the cancer cell. The disruption may affect one or more signaling or expression events. For example the encoded oncology-related polypeptide may act upstream of a transcription factor known to induce or enhance the expression of genes associated with a cancer. Delivery of the signal-sensor polynucleotide encoding the oncology-related polypeptide which inhibits such a transcription factor (either by binding or sequestration or degradation) would thereby alter the transcriptome of the cancer cell and have a therapeutic benefit. One such transcription factor is HIF-1alpha. A signal-sensor polynucleotide encoding a protein which is capable of binding HIF-1alpha or whose expression results in lower HIF-1alpha, would effectively turn down HIF-1alpha regulated genes, e.g., VEGFA or SLC2A1, and destabilize the cancer.

In one embodiment, the profile of the cancer may be evaluated before the signal-sensor polynucleotide is selected. Such profiling data would inform the selection of which oncology-related polypeptide to be delivered. The profile of gene expression, categorized by hallmark class such as apoptosis, replicative capacity or metabolic signature would allow dynamic instability scoring for a polypeptide and an optimization of therapeutic window for the signal-sensor polynucleotide. As used herein, a “dynamic instability index” refers to a dose of signal-sensor polynucleotide sufficient to induce 50% increase of the oncology-related target protein in vitro in a cancer cell as compared to a normal matched cell.

Profiling may also be done within hallmark classes such as the distinction between caspase-dependent and caspase independent gene expression for the apoptosis class. Alternatively, profiling could be conducted across classes such as gene profiling of apoptosis, senescence (replicative capacity), and metabolic classes.

In one embodiment, the signal-sensor polynucleotides described herein may be used to reduce the expression and/or amount of a polypeptide in a cell. As a non-limiting example, MYC inhibitor A, MYC inhibitor B, MYC inhibitor C or MYC inhibitor D may be used on Hep3B cells in order to determine the potency of MYC inhibitor A, MYC inhibitor B, MYC inhibitor C or MYC inhibitor D at various concentrations (see e.g., Example 55).

In one embodiment, the signal-sensor polynucleotides described herein may direct either cytotoxic or cytoprotective therapeutic benefit to specific cells, e.g., normal vs. cancerous.

In one embodiment signal-sensor polynucleotides would not only encode an oncology-related polypeptide but also a sensor sequence. Sensor sequences include, for example, microRNA binding sites, transcription factor binding sites, artificial binding sites engineered to act as pseudo-receptors for endogenous nucleic acid binding molecules. A “sensor region” is a region of linked nucleosides of the signal-sensor polynucleotide comprising at least one sensor sequence. The signal-sensor polynucleotides of the present invention may have one or more sensor regions.

In one embodiment, one or more sensor regions may be located in the first flanking region. As a non-limiting example, the sensor region in the first flanking region may comprise at least one sensor sequence. The sensor sequence may be, but is not limited to, mir-122, mir-142-3p, mir-142-5p, mir-146, fragments or variants thereof. As another non-limiting example, the sensor region in the first flanking region may comprise at least one sensor sequence such as a mir-122 sequence. The mir-122 sequence may be, but is not limited to, a mir-122 binding site, mir-122 seed sequence, mir-122 binding site without the seed sequence or a combination thereof.

In another embodiment, one or more sensor regions may be located in the second flanking region. As a non-limiting example, the sensor region in the second flanking region may include a sensor sequence such as mir-122, mir-142-3p, mir-142-5p, mir-146, fragments or variants thereof. As another non-limiting example, the sensor region in the second flanking region may include three sensor sequences. The sensor sequences may be, but are not limited to, mir-122 sequences such as mir-122 binding sites, mir-122 seed sequences, mir-122 binding sites without the seed sequence or a combination thereof. As yet another non-limiting example, the sensor region in the second flanking region is located in the 3′UTR and the sensor region may include a sensor sequence which is a mir-122 sequence. The mir-122 sequence may be, but is not limited to, a mir-122 binding site, mir-122 seed sequence, mir-122 binding site without the seed sequence or a combination thereof.

In one embodiment, two or more sensor regions may be located in the same region of the signal-sensor polynucleotide such as, but not limited to, a first region first region of linked nucleotides, the first flanking region and/or the second flanking region. As a non-limiting example, the two or more sensor regions are located in the second flanking region. As yet another non-limiting example, three sensor regions are located in the 3′ UTR in the second flanking region. The three sensor regions may include, mir-122 binding sites, mir-122 seed sequences, mir-122 binding sites without the seed sequence or a combination thereof.

In another embodiment, two or more sensor regions may be located in different regions of the signal-sensor polynucleotide such as, but not limited to, the first region of linked nucleotides, the first flanking region and/or the second flanking region. As a non-limiting example, a first sensor region is located in the first flanking region and a second sensor region is located in the second flanking region. The sensor regions may comprise the same sensor sequence or different sensor sequences.

In one embodiment, a start codon is located within a sensor region.

In one embodiment, a sensor region may comprise two or more sensor sequences. The sensor sequences may be the same or different.

In one embodiment, the sensor region may comprise two or more sensor sequence which are different from each other but they may be based on the same mir binding site. As a non-limiting example, the sensor region may include at least one miR binding site sequence and at least one mir binding site sequence with the seed removed. As another non-limiting example, the sensor region may include at least one miR binding site sequence and at least one miR seed sequence. As yet another non-limiting example, the sensor region may include at least one miR binding site sequence with the seed removed and at least one miR seed sequence.

In another embodiment, the sensor region may comprise two or more sensor sequences which are in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than three times. In these patterns, each letter, A, B, or C represent a different miR sequence.

In yet another embodiment, the signal-sensor polynucleotide may include two or more sensor regions with each sensor region having one or more sensor sequences. As a non-limiting example, the sensor sequences may be in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than three times in each of the sensor regions. As another non-limiting example, the sensor sequences may be in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than three times across the entire signal-sensor polynucleotide. In these patterns, each letter, A, B, or C represent a different miR sequence. As a non-limiting example, the first sensor region may have sensor sequences in the pattern ABA and the second sensor region may have sensor sequences in the pattern BAB so the overall pattern of the sensor sequences in the signal-sensor polynucleotide is ABABAB. As another non-limiting example, the first sensor region may have sensor sequences AA, the second sensor region may have sensor sequences BB, the third sensor region may have sensor sequences AA and the fourth sensor region may have sensor sequences BB so the overall pattern of the sensor sequences in the signal-sensor polynucleotide is AABBAABB.

The sensor sequences in the signal-sensor polynucleotides of the present invention may include one or more regulatory sequences in the 3-UTR and/or 5′UTR of natural mRNAs, which regulate mRNA stability and translation in different tissues and cells. Such cis-regulatory elements may include, but are not limited to, Cis-RNP (Ribonucleoprotein)/RBP (RNA binding protein) regulatory elements, AU-rich element AUE, structured stem-loop, constitutive decay elements (CDEs), GC-richness and other structured mRNA motifs (Parker B J et al., Genome Research, 2011, 21, 1929-1943, which is herein incorporated by reference in its entirety.). For example, CDEs are a class of regulatory motifs that mediate mRNA degradation through their interaction with Roquin proteins. In particular, CDEs are found in many mRNAs that encode regulators of development and inflammation to limit cytokine production in macrophage (Leppek K et al., Cell, 2013, 153, 869-881, which is herein incorporated by reference in its entirety.).

In one embodiment, a particular CDE can be introduced to the signal-sensor polynucleotide when the degradation of polypeptides in a cell or tissue is desired. A particular CDE can also be removed from the signal-sensor polynucleotide in order to maintain a more stable mRNA in a cell or tissue for sustaining protein expression.

In one embodiment, microRNA (miRNA) profiling of the cancer cells or tissues may be conducted to determine the presence or absence of miRNA in the cells or tissues to determine the appropriate microRNA to use as sensor sequences in the signal sensor polynucleotides.

MicroRNA gene regulation may be influenced by the sequence surrounding the microRNA such as, but not limited to, the species of the surrounding sequence, the type of sequence (e.g., heterologous, hom*ologous and artificial), regulatory elements in the surrounding sequence and/or structural elements in the surrounding sequence. The microRNA may be influenced by the 5′UTR and/or the 3′UTR. As a non-limiting example, a non-human 3′UTR may increase the regulatory effect of the microRNA sequence on the expression of a polypeptide of interest compared to a human 3′UTR of the same sequence type.

Other regulatory elements and/or structural elements of the 5′-UTR can influence microRNA mediated gene regulation. One such example is a structured IRES (Internal Ribosome Entry Site) in the 5′UTR, which is necessary for the binding of translational elongation factors to initiate protein translation. EIF4A2 binding to this secondarily structured element in the 5′UTR is necessary for microRNA mediated gene expression (Meijer H A et al., Science, 2013, 340, 82-85, herein incorporated by reference in its entirety). The sensor-signal polynucleotide can further be modified to include this structured 5′-UTR in order to enhance microRNA mediated gene regulation.

At least one microRNA site can be engineered into the 3′ UTR of the signal-sensor polynucleotides of the present invention. In this context, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten or more microRNA sites may be engineered into the 3′ UTR of the signal-sensor polynucleotides of the present invention. In one embodiment, the microRNA sites incorporated into the signal-sensor polynucleotides may be the same or may be different microRNA sites. In another embodiment, the microRNA sites incorporated into the signal-sensor polynucleotides may target the same or different tissues in the body. As a non-limiting example, through the introduction of tissue-, cell-type-, or disease-specific microRNA binding sites in the 3′ UTR of a signal-sensor polynucleotide, the degree of expression in specific cell types (e.g. hepatocytes, myeloid cells, endothelial cells, cancer cells, etc.) can be reduced.

In one embodiment, a microRNA site can be engineered near the 5′ terminus of the 3′UTR, about halfway between the 5′ terminus and 3′ terminus of the 3′UTR and/or near the 3′ terminus of the 3′UTR. As a non-limiting example, a microRNA site may be engineered near the 5′ terminus of the 3′UTR and about halfway between the 5′ terminus and 3′ terminus of the 3′UTR. As another non-limiting example, a microRNA site may be engineered near the 3′ terminus of the 3′UTR and about halfway between the 5′ terminus and 3′ terminus of the 3′UTR. As yet another non-limiting example, a microRNA site may be engineered near the 5′ terminus of the 3′UTR and near the 3′ terminus of the 3′UTR.

In another embodiment, a 3′UTR can comprise 4 microRNA sites. The microRNA sites may be complete microRNA binding sites, microRNA seed sequences and/or microRNA binding site sequences without the seed sequence.

In one embodiment, a signal-sensor polynucleotide may be engineered to include microRNA sites which are expressed in different tissues of a subject. As a non-limiting example, a signal-sensor polynucleotide of the present invention may be engineered to include miR-192 and miR-122 to regulate expression of the signal-sensor polynucleotide in the liver and kidneys of a subject. In another embodiment, a signal-sensor polynucleotide may be engineered to include more than one microRNA sites for the same tissue. For example a signal-sensor polynucleotide of the present invention may be engineered to include miR-17-92 and miR-126 to regulate expression of the signal-sensor polynucleotide in endothelial cells of a subject.

In one embodiment, the therapeutic window and or differential expression associated with the oncology-related polypeptide encoded by the signal-sensor polynucleotide of the invention may be altered. For example, signal-sensor polynucleotides may be designed whereby a death signal is more highly expressed in cancer cells (or a survival signal in a normal cell) by virtue of the miRNA signature of those cells. Where a cancer cell expresses a lower level of a particular miRNA, the signal-sensor polynucleotide encoding the binding site for that miRNA (or miRNAs) would be more highly expressed. Hence, the oncology-related polypeptide encoded by the signal-sensor polynucleotide is selected as a protein which triggers or induces cell death. Neighboring noncancer cells, harboring a higher expression of the same miRNA would be less affected by the encoded death signal as the signal-sensor polynucleotide would be expressed at a lower level due to the affects of the miRNA binding to the binding site or “sensor” encoded in the 3′UTR. Conversely, cell survival or cytoprotective signals may be delivered to tissues containing cancer and non cancerous cells where a miRNA has a higher expression in the cancer cells—the result being a lower survival signal to the cancer cell and a larger survival signature to the normal cell. Multiple signal-sensor polynucleotides may be designed and administered having different signals according to the previous paradigm.

In one embodiment, the expression of a signal-sensor polynucleotide may be controlled by incorporating at least one sensor sequence in the signal-sensor polynucleotide and formulating the signal-sensor polynucleotide. As a non-limiting example, a polynucleotide may be targeted to an orthotopic tumor by having a polynucleotide incorporating a miR-122 binding site and formulated in a lipid nanoparticle comprising the cationic lipid DLin-KC2-DMA (see e.g., the experiments described in Example 56A and 56B).

Through an understanding of the expression patterns of microRNA in different cell types, signal-sensor polynucleotides can be engineered for more targeted expression in specific cell types or only under specific biological conditions. Through introduction of tissue-specific microRNA binding sites, signal-sensor polynucleotides could be designed that would be optimal for protein expression in a tissue or in the context of a biological condition such as cancer.

Transfection experiments can be conducted in relevant cell lines, using engineered signal-sensor polynucleotides and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different microRNA binding site-engineering nucleic acids or signal-sensor polynucleotides and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hr, 12 hr, 24 hr, 48 hr, 72 hr and 7 days post-transfection. In vivo experiments can also be conducted using microRNA-binding site-engineered molecules to examine changes in tissue-specific expression of formulated signal-sensor polynucleotides.

In one embodiment, the signal-sensor polynucleotides of the invention may include at least one microRNA in order to dampen the antigen presentation by antigen presenting cells. The microRNA may be the complete microRNA sequence, the microRNA seed sequence, the microRNA sequence without the seed or a combination thereof. As a non-limiting example, the microRNA incorporated into the signal-sensor polynucleotide may be specific to the hematopoietic system. As another non-limiting example, the microRNA incorporated into the signal-sensor polynucleotides of the invention to dampen antigen presentation is miR-142-3p.

In one embodiment, the signal-sensor polynucleotides of the invention may include at least one microRNA in order to dampen expression of the encoded polypeptide in a cell of interest. As a non-limiting example, the signal-sensor polynucleotides of the invention may include at least one miR-122 binding site in order to dampen expression of an encoded polypeptide of interest in the liver. As another non-limiting example, the signal-sensor polynucleotides of the invention may include at least one miR-142-3p binding site, miR-142-3p seed sequence, miR-142-3p binding site without the seed, miR-142-5p binding site, miR-142-5p seed sequence, miR-142-5p binding site without the seed, miR-146 binding site, miR-146 seed sequence and/or miR-146 binding site without the seed sequence (see e.g., the experiment outlined in Example 47 and Example 60).

According to the present invention, the signal-sensor polynucleotides described herein may be modified as to avoid the deficiencies of other polypeptide-encoding molecules of the art. Hence, in this embodiment the signal-sensor polynucleotides are referred to as modified signal-sensor polynucleotides or primary constructs, modified mRNA or mmRNA.

Provided herein, in part, are signal-sensor polynucleotides, primary constructs and/or mmRNA encoding oncology-related polypeptides of interest which have been designed to improve one or more of the stability and/or clearance in tissues, receptor uptake and/or kinetics, cellular access by the compositions, engagement with translational machinery, mRNA half-life, translation efficiency, immune evasion, protein production capacity, secretion efficiency (when applicable), accessibility to circulation, protein half-life and/or modulation of a cell's status, function and/or activity.

The present invention provides nucleic acid molecules, specifically signal-sensor polynucleotides, primary constructs and/or mmRNA which encode one or more oncology-related polypeptides of interest. Specifically the invention contemplates signal-sensor polynucleotides which are useful in cancer or cancer related diseases, disorders. As used herein, “signal-sensor polynucleotides” are nucleic acid transcripts which encode one or more oncology-related polypeptides of interest that, when translated, delivers a “signal” to the cell (cancer or noncancerous) which results in the therapeutic benefit to the organism of either being detrimental to the cancer cell or beneficial to normal cells or both detrimental to cancer cells and advantageous to normal cells. The signal-sensor polynucleotides may optionally further comprise a sequence (translatable or not) which “senses” the microenvironment of the polynucleotide and alters (a) the function or phenotypic outcome associated with the peptide or protein which is translated, (b) the expression level of the signal-sensor polynucleotide, and/or both.

The term “nucleic acid,” in its broadest sense, includes any compound and/or substance that comprise a polymer of nucleotides. These polymers are often referred to as polynucleotides. Exemplary nucleic acids or polynucleotides of the invention include, but are not limited to, ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs, including LNA having a β-D-ribo configuration, α-LNA having an α-L-ribo configuration (a diastereomer of LNA), 2′-amino-LNA having a 2′-amino functionalization, and 2′-amino-α-LNA having a 2′-amino functionalization) or hybrids thereof.

In preferred embodiments, the signal-sensor polynucleotide or nucleic acid molecule is a messenger RNA (mRNA). As used herein, the term “messenger RNA” (mRNA) refers to any polynucleotide which encodes a polypeptide of interest and which is capable of being translated to produce the encoded polypeptide of interest in vitro, in vivo, in situ or ex vivo. Signal-sensor polynucleotides of the invention may be mRNA or any nucleic acid molecule and may or may not be chemically modified.

Traditionally, the basic components of an mRNA molecule include at least a coding region, a 5′UTR, a 3′UTR, a 5′ cap and a poly-A tail. Building on this wild type modular structure, the present invention expands the scope of functionality of traditional mRNA molecules by providing signal-sensor polynucleotides or primary RNA constructs which maintain a modular organization, but which comprise one or more structural and/or chemical modifications or alterations which impart useful properties to the polynucleotide including, in some embodiments, the lack of a substantial induction of the innate immune response of a cell into which the signal-sensor polynucleotide is introduced. As such, modified mRNA molecules of the present invention, which may be synthetic, are termed “mmRNA.” As used herein, a “structural” feature or modification is one in which two or more linked nucleotides are inserted, deleted, duplicated, inverted or randomized in a signal-sensor polynucleotide, primary construct or mmRNA without significant chemical modification to the nucleotides themselves.

Because chemical bonds will necessarily be broken and reformed to effect a structural modification, structural modifications are of a chemical nature and hence are chemical modifications. However, structural modifications will result in a different sequence of nucleotides. For example, the polynucleotide “ATCG” may be chemically modified to “AT-5meC-G”. The same polynucleotide may be structurally modified from “ATCG” to “ATCCCG”. Here, the dinucleotide “CC” has been inserted, resulting in a structural modification to the polynucleotide.

Signal-Sensor Polynucleotide, Primary Construct or mmRNA Architecture

The signal-sensor polynucleotides of the present invention are distinguished from wild type mRNA in their functional and/or structural design features which serve to, as evidenced herein, overcome existing problems of effective polypeptide production using nucleic acid-based therapeutics.

FIG. 1 shows a representative signal-sensor primary construct 100 of the present invention. As used herein, the term “primary construct” or “primary mRNA construct” refers to a signal-sensor polynucleotide transcript which encodes one or more polypeptides of interest and which retains sufficient structural and/or chemical features to allow the polypeptide of interest encoded therein to be translated. Signal-sensor primary constructs may be polynucleotides of the invention. When structurally or chemically modified, the signal-sensor primary construct may be referred to as a mmRNA.

Returning to FIG. 1, the primary construct 100 here contains a first region of linked nucleotides 102 that is flanked by a first flanking region 104 and a second flaking region 106. As used herein, the “first region” may be referred to as a “coding region” or “region encoding” or simply the “first region.” This first region may include, but is not limited to, the encoded oncology-related polypeptide of interest. The oncology-related polypeptide of interest may comprise at its 5′ terminus one or more signal peptide sequences encoded by a signal peptide sequence region 103. The flanking region 104 may comprise a region of linked nucleotides comprising one or more complete or incomplete 5′ UTRs sequences. The flanking region 104 may also comprise a 5′ terminal cap 108. The second flanking region 106 may comprise a region of linked nucleotides comprising one or more complete or incomplete 3′ UTRs. The flanking region 106 may also comprise a 3′ tailing sequence 110 and a 3′UTR 120.

Bridging the 5′ terminus of the first region 102 and the first flanking region 104 is a first operational region 105. Traditionally this operational region comprises a start codon. The operational region may alternatively comprise any translation initiation sequence or signal including a start codon.

Bridging the 3′ terminus of the first region 102 and the second flanking region 106 is a second operational region 107. Traditionally this operational region comprises a stop codon. The operational region may alternatively comprise any translation initiation sequence or signal including a stop codon. According to the present invention, multiple serial stop codons may also be used. In one embodiment, the operation region of the present invention may comprise two stop codons. The first stop codon may be “TGA” and the second stop codon may be selected from the group consisting of “TAA,” “TGA” and “TAG.” The operation region may further comprise three stop codons. The third stop codon may be selected from the group consisting of “TAA,” “TGA” and “TAG.”

Turning to FIG. 2, the 3′UTR 120 of the second flanking region 106 may comprise one or more sensor sequences 130. A region comprising at least one sensor sequence is referred to as a “sensor region.” These sensor sequences as discussed herein operate as pseudo-receptors (or binding sites) for ligands of the local microenvironment of the primary construct or signal-sensor polynucleotide. For example, microRNA binding sites or miRNA seeds may be used as sensors such that they function as pseudoreceptors for any microRNAs present in the environment of the polynucleotide.

Generally, the shortest length of the first region of the signal-sensor primary construct of the present invention can be the length of a nucleic acid sequence that is sufficient to encode for a dipeptide, a tripeptide, a tetrapeptide, a pentapeptide, a hexapeptide, a heptapeptide, an octapeptide, a nonapeptide, or a decapeptide. In another embodiment, the length may be sufficient to encode a peptide of 2-30 amino acids, e.g. 5-30, 10-30, 2-25, 5-25, 10-25, or 10-20 amino acids. The length may be sufficient to encode for a peptide of at least 11, 12, 13, 14, 15, 17, 20, 25 or 30 amino acids, or a peptide that is no longer than 40 amino acids, e.g. no longer than 35, 30, 25, 20, 17, 15, 14, 13, 12, 11 or 10 amino acids. Examples of dipeptides that the polynucleotide sequences can encode or include, but are not limited to, carnosine and anserine.

Generally, the length of the first region encoding the oncology-related polypeptide of interest of the present invention is greater than about 30 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000, 4,000, 5,000, 6,000, 7,000, 8,000, 9,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000 or up to and including 100,000 nucleotides). As used herein, the “first region” may be referred to as a “coding region” or “region encoding” or simply the “first region.”

In some embodiments, the signal-sensor polynucleotide, primary construct, or mmRNA includes from about 30 to about 100,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 1,000, from 30 to 1,500, from 30 to 3,000, from 30 to 5,000, from 30 to 7,000, from 30 to 10,000, from 30 to 25,000, from 30 to 50,000, from 30 to 70,000, from 100 to 250, from 100 to 500, from 100 to 1,000, from 100 to 1,500, from 100 to 3,000, from 100 to 5,000, from 100 to 7,000, from 100 to 10,000, from 100 to 25,000, from 100 to 50,000, from 100 to 70,000, from 100 to 100,000, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 3,000, from 500 to 5,000, from 500 to 7,000, from 500 to 10,000, from 500 to 25,000, from 500 to 50,000, from 500 to 70,000, from 500 to 100,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 3,000, from 1,000 to 5,000, from 1,000 to 7,000, from 1,000 to 10,000, from 1,000 to 25,000, from 1,000 to 50,000, from 1,000 to 70,000, from 1,000 to 100,000, from 1,500 to 3,000, from 1,500 to 5,000, from 1,500 to 7,000, from 1,500 to 10,000, from 1,500 to 25,000, from 1,500 to 50,000, from 1,500 to 70,000, from 1,500 to 100,000, from 2,000 to 3,000, from 2,000 to 5,000, from 2,000 to 7,000, from 2,000 to 10,000, from 2,000 to 25,000, from 2,000 to 50,000, from 2,000 to 70,000, and from 2,000 to 100,000).

According to the present invention, the first and second flanking regions may range independently from 15-1,000 nucleotides in length (e.g., greater than 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, and 900 nucleotides or at least 30, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, and 1,000 nucleotides).

According to the present invention, the tailing sequence may range from absent to 500 nucleotides in length (e.g., at least 60, 70, 80, 90, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, or 500 nucleotides). Where the tailing region is a polyA tail, the length may be determined in units of or as a function of polyA binding protein binding. In this embodiment, the polyA tail is long enough to bind at least 4 monomers of polyA binding protein. PolyA binding protein monomers bind to stretches of approximately 38 nucleotides. As such, it has been observed that polyA tails of about 80 nucleotides and 160 nucleotides are functional.

According to the present invention, the capping region may comprise a single cap or a series of nucleotides forming the cap. In this embodiment the capping region may be from 1 to 10, e.g. 2-9, 3-8, 4-7, 1-5, 5-10, or at least 2, or 10 or fewer nucleotides in length. In some embodiments, the cap is absent.

According to the present invention, the first and second operational regions may range from 3 to 40, e.g., 5-30, 10-20, 15, or at least 4, or 30 or fewer nucleotides in length and may comprise, in addition to a start and/or stop codon, one or more signal and/or restriction sequences.

Cyclic Signal-Sensor Polynucleotides

According to the present invention, a signal-sensor primary construct or mmRNA may be cyclized, or concatemerized, to generate a translation competent molecule to assist interactions between poly-A binding proteins and 5′-end binding proteins. The mechanism of cyclization or concatemerization may occur through at least 3 different routes: 1) chemical, 2) enzymatic, and 3) ribozyme catalyzed. The newly formed 5′/3′-linkage may be intramolecular or intermolecular.

In the first route, the 5′-end and the 3′-end of the nucleic acid may contain chemically reactive groups that, when close together, form a new covalent linkage between the 5′-end and the 3′-end of the molecule. The 5′-end may contain an NHS-ester reactive group and the 3′-end may contain a 3′-amino-terminated nucleotide such that in an organic solvent the 3′-amino-terminated nucleotide on the 3′-end of a synthetic mRNA molecule will undergo a nucleophilic attack on the 5′-NHS-ester moiety forming a new 5′-/3′-amide bond.

In the second route, T4 RNA ligase may be used to enzymatically link a 5′-phosphorylated nucleic acid molecule to the 3′-hydroxyl group of a nucleic acid forming a new phosphoro diester linkage. In an example reaction, 1 μg of a nucleic acid molecule is incubated at 37° C. for 1 hour with 1-10 units of T4 RNA ligase (New England Biolabs, Ipswich, Mass.) according to the manufacturer's protocol. The ligation reaction may occur in the presence of a split oligonucleotide capable of base-pairing with both the 5′- and 3′-region in juxtaposition to assist the enzymatic ligation reaction.

In the third route, either the 5′- or 3′-end of the cDNA template encodes a ligase ribozyme sequence such that during in vitro transcription, the resultant nucleic acid molecule can contain an active ribozyme sequence capable of ligating the 5′-end of a nucleic acid molecule to the 3′-end of a nucleic acid molecule. The ligase ribozyme may be derived from the Group I Intron, Group I Intron, Hepatitis Delta Virus, Hairpin ribozyme or may be selected by SELEX (systematic evolution of ligands by exponential enrichment). The ribozyme ligase reaction may take 1 to 24 hours at temperatures between 0 and 37° C.

Signal-Sensor Polynucleotide Multimers

According to the present invention, multiple distinct signal-sensor polynucleotides, primary constructs or mmRNA may be linked together through the 3′-end using nucleotides which are modified at the 3′-terminus. Chemical conjugation may be used to control the stoichiometry of delivery into cells. For example, the glyoxylate cycle enzymes, isocitrate lyase and malate synthase, may be supplied into HepG2 cells at a 1:1 ratio to alter cellular fatty acid metabolism. This ratio may be controlled by chemically linking signal-sensor polynucleotides, primary constructs or mmRNA using a 3′-azido terminated nucleotide on one signal-sensor polynucleotide, primary construct or mmRNA species and a C5-ethynyl or alkynyl-containing nucleotide on the opposite signal-sensor polynucleotide, primary construct or mmRNA species. The modified nucleotide is added post-transcriptionally using terminal transferase (New England Biolabs, Ipswich, Mass.) according to the manufacturer's protocol. After the addition of the 3′-modified nucleotide, the two signal-sensor polynucleotide, primary construct or mmRNA species may be combined in an aqueous solution, in the presence or absence of copper, to form a new covalent linkage via a click chemistry mechanism as described in the literature.

In another example, more than two signal-sensor polynucleotides may be linked together using a functionalized linker molecule. For example, a functionalized saccharide molecule may be chemically modified to contain multiple chemical reactive groups (SH—, NH2—, N3, etc. . . . ) to react with the cognate moiety on a 3′-functionalized signal-sensor polynucleotide molecule (i.e., a 3′-maleimide ester, 3′-NHS-ester, alkynyl). The number of reactive groups on the modified saccharide can be controlled in a stoichiometric fashion to directly control the stoichiometric ratio of conjugated signal-sensor polynucleotide, primary construct or mmRNA.

Signal-Sensor Polynucleotide Conjugates and Combinations

In order to further enhance oncology-related protein production, signal-sensor polynucleotide primary constructs or mmRNA of the present invention can be designed to be conjugated to other polynucleotides, oncology-related polypeptides, dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases, proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell, hormones and hormone receptors, non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, or a drug.

Conjugation may result in increased stability and/or half life and may be particularly useful in targeting the signal-sensor polynucleotides, primary constructs or mmRNA to specific sites in the cell, tissue or organism.

According to the present invention, the signal-sensor polynucleotide mmRNA or primary constructs may be administered with, or further encode one or more of RNAi agents, siRNAs, shRNAs, miRNAs, miRNA binding sites, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, aptamers or vectors, and the like.

In one embodiment, the signal-sensor polynucleotides described herein may be conjugated with a moiety to target various cancer cells such as, but not limited to, the moieties described in US Patent Application No. US20130216561, the contents of which are herein incorporated by reference in its entirety. The linkage between the signal-sensor polynucleotides and the cancer targeting moiety may be an acid cleavable linkage that can increase the efficacy of the conjugate such as, but not limited to, the linkages described in US Patent Application No. US20130216561, the contents of which are herein incorporated by reference in its entirety.

Bifunctional Signal-Sensor Polynucleotide

In one embodiment of the invention are bifunctional signal-sensor polynucleotides (e.g., bifunctional primary constructs or bifunctional mmRNA). As the name implies, bifunctional signal-sensor polynucleotides are those having or capable of at least two functions. These molecules may also by convention be referred to as multifunctional.

The multiple functionalities of bifunctional signal-sensor polynucleotides may be encoded by the RNA (the function may not manifest until the encoded product is translated) or may be a property of the polynucleotide itself. It may be structural or chemical. Bifunctional modified signal-sensor polynucleotides may comprise a function that is covalently or electrostatically associated with the polynucleotides. Further, the two functions may be provided in the context of a complex of a signal-sensor polynucleotide and another molecule.

Bifunctional signal-sensor polynucleotides may encode oncology-related peptides which are anti-proliferative. These peptides may be linear, cyclic, constrained or random coil. They may function as aptamers, signaling molecules, ligands or mimics or mimetics thereof. Anti-proliferative peptides may, as translated, be from 3 to 50 amino acids in length. They may be 5-40, 10-30, or approximately 15 amino acids long. They may be single chain, multichain or branched and may form complexes, aggregates or any multi-unit structure once translated.

Noncoding Signal-Sensor Polynucleotides

As described herein, provided are signal-sensor polynucleotides and primary constructs having sequences that are partially or substantially not translatable, e.g., having a noncoding region. Such noncoding region may be the “first region” of the signal-sensor primary construct. Alternatively, the noncoding region may be a region other than the first region. Such molecules are generally not translated, but can exert an effect on protein production by one or more of binding to and sequestering one or more translational machinery components such as a ribosomal protein or a transfer RNA (tRNA), thereby effectively reducing protein expression in the cell or modulating one or more pathways or cascades in a cell which in turn alters protein levels. The signal-sensor polynucleotide and/or primary construct may contain or encode one or more long noncoding RNA (lncRNA, or lincRNA) or portion thereof, a small nucleolar RNA (sno-RNA), micro RNA (miRNA), small interfering RNA (siRNA) or Piwi-interacting RNA (piRNA).

Auxotrophic Signal-Sensor Polynucleotides

In one embodiment, the signal-sensor polynucleotides of the present invention may be auxotrophic. As used herein, the term “auxotrophic” refers to signal-sensor polynucleotides that comprise at least one feature that triggers, facilitates or induces the degradation or inactivation of the itself in response to spatial or temporal cues such that oncology-related protein expression is substantially prevented or reduced. Such spatial or temporal cues include the location of the signal-sensor polynucleotide to be translated such as a particular tissue or organ or cellular environment. Also contemplated are cues involving temperature, pH, ionic strength, moisture content, and the like.

In one embodiment, the feature is located in a terminal region of the signal-sensor polynucleotides of the present invention. As a non-limiting example, the auxotrophic mRNA may contain a miR binding site in the terminal region which binds to a miR expressed in a selected tissue so that the expression of the auxotrophic mRNA is substantially prevented or reduced in the selected tissue. To this end and for example, an auxotrophic mRNA containing a miR-122 binding site will not produce protein if localized to the liver since miR-122 is expressed in the liver and binding of the miR would effectuate destruction of the auxotrophic mRNA. As a non-limiting example, HEK293 cells do not express miR-122 so there would be little to no downregulation of a signal-sensor polynucleotide having a miR-122 sequence in HEK293 but for hepatocytes which do expression miR-122 there would be a downregulation of a signal-sensor polynucleotide having a miR-122 sequence in hepatocytes (see e.g., the study outlined Example 19). As another non-limiting example, the miR-122 level can be measured in HeLa cells, primary human hepatocytes and primary rat hepatocytes prior to administration with a signal-sensor polynucleotide encoding having at least one miR-122 binding site, miR-122 binding site without the seed sequence or a miR-122 binding site After administration the expression of the signal-sensor polynucleotide can be measured to determine the dampening effect of the miR-122 in the signal-sensor polynucleotide (see e.g., the studies outlined in Examples 41, 42, 43 57, 58 and 59). As yet another non-limiting example, the effectiveness of the miR-122 binding site, miR-122 seed or the miR-122 binding site without the seed in different 3′UTRs may be evaluated in order to determine the proper UTR for the desired outcome such as, but not limited to, the highest dampening effect (see e.g., the study outlined in Example 46).

In one embodiment, the degradation or inactivation of auxotrophic mRNA may comprise a feature responsive to a change in pH. As a non-limiting example, the auxotrophic mRNA may be triggered in an environment having a pH of between pH 4.5 to 8.0 such as at a pH of 5.0 to 6.0 or a pH of 6.0 to 6.5. The change in pH may be a change of 0.1 unit, 0.2 units, 0.3 units, 0.4 units, 0.5 units, 0.6 units, 0.7 units, 0.8 units, 0.9 units, 1.0 units, 1.1 units, 1.2 units, 1.3 units, 1.4 units, 1.5 units, 1.6 units, 1.7 units, 1.8 units, 1.9 units, 2.0 units, 2.1 units, 2.2 units, 2.3 units, 2.4 units, 2.5 units, 2.6 units, 2.7 units, 2.8 units, 2.9 units, 3.0 units, 3.1 units, 3.2 units, 3.3 units, 3.4 units, 3.5 units, 3.6 units, 3.7 units, 3.8 units, 3.9 units, 4.0 units or more.

In another embodiment, the degradation or inactivation of auxotrophic mRNA may be triggered or induced by changes in temperature. As a non-limiting example, a change of temperature from room temperature to body temperature. The change of temperature may be less than 1° C., less than 5° C., less than 10° C., less than 15° C., less than 20° C., less than 25° C. or more than 25° C.

In yet another embodiment, the degradation or inactivation of auxotrophic mRNA may be triggered or induced by a change in the levels of ions in the subject. The ions may be cations or anions such as, but not limited to, sodium ions, potassium ions, chloride ions, calcium ions, magnesium ions and/or phosphate ions.

Oncology-Related Polypeptides of Interest

According to the present invention, the signal-sensor primary construct is designed to encode one or more oncology-related polypeptides of interest or fragments thereof. An oncology-related polypeptide of interest may include, but is not limited to, whole polypeptides, a plurality of polypeptides or fragments of polypeptides, which independently may be encoded by one or more nucleic acids, a plurality of nucleic acids, fragments of nucleic acids or variants of any of the aforementioned. As used herein, the term “oncology-related polypeptides of interest” refers to any polypeptide which is selected to be encoded in the signal-sensor primary construct of the present invention. As used herein, “polypeptide” means a polymer of amino acid residues (natural or unnatural) linked together most often by peptide bonds. The term, as used herein, refers to proteins, polypeptides, and peptides of any size, structure, or function. In some instances the polypeptide encoded is smaller than about 50 amino acids and the polypeptide is then termed a peptide. If the polypeptide is a peptide, it will be at least about 2, 3, 4, or at least 5 amino acid residues long. Thus, polypeptides include gene products, naturally occurring polypeptides, synthetic polypeptides, hom*ologs, orthologs, paralogs, fragments and other equivalents, variants, and analogs of the foregoing. A polypeptide may be a single molecule or may be a multi-molecular complex such as a dimer, trimer or tetramer. They may also comprise single chain or multichain polypeptides such as antibodies or insulin and may be associated or linked. Most commonly disulfide linkages are found in multichain polypeptides. The term polypeptide may also apply to amino acid polymers in which one or more amino acid residues are an artificial chemical analogue of a corresponding naturally occurring amino acid.

The term “polypeptide variant” refers to molecules which differ in their amino acid sequence from a native or reference sequence. The amino acid sequence variants may possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence, as compared to a native or reference sequence. Ordinarily, variants will possess at least about 50% identity (hom*ology) to a native or reference sequence, and preferably, they will be at least about 80%, more preferably at least about 90% identical (hom*ologous) to a native or reference sequence.

In some embodiments “variant mimics” are provided. As used herein, the term “variant mimic” is one which contains one or more amino acids which would mimic an activated sequence. For example, glutamate may serve as a mimic for phosphoro-threonine and/or phosphoro-serine. Alternatively, variant mimics may result in deactivation or in an inactivated product containing the mimic, e.g., phenylalanine may act as an inactivating substitution for tyrosine; or alanine may act as an inactivating substitution for serine.

“hom*ology” as it applies to amino acid sequences is defined as the percentage of residues in the candidate amino acid sequence that are identical with the residues in the amino acid sequence of a second sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent hom*ology. Methods and computer programs for the alignment are well known in the art. It is understood that hom*ology depends on a calculation of percent identity but may differ in value due to gaps and penalties introduced in the calculation.

By “hom*ologs” as it applies to polypeptide sequences means the corresponding sequence of other species having substantial identity to a second sequence of a second species.

“Analogs” is meant to include polypeptide variants which differ by one or more amino acid alterations, e.g., substitutions, additions or deletions of amino acid residues that still maintain one or more of the properties of the parent or starting polypeptide.

The present invention contemplates several types of compositions which are polypeptide based including variants and derivatives. These include substitutional, insertional, deletion and covalent variants and derivatives. The term “derivative” is used synonymously with the term “variant” but generally refers to a molecule that has been modified and/or changed in any way relative to a reference molecule or starting molecule.

As such, signal-sensor polynucleotides encoding oncology-related polypeptides containing substitutions, insertions and/or additions, deletions and covalent modifications with respect to reference sequences, in particular the oncology-related polypeptide sequences disclosed herein, are included within the scope of this invention. For example, sequence tags or amino acids, such as one or more lysines, can be added to the peptide sequences of the invention (e.g., at the N-terminal or C-terminal ends). Sequence tags can be used for peptide purification or localization. Lysines can be used to increase peptide solubility or to allow for biotinylation. Alternatively, amino acid residues located at the carboxy and amino terminal regions of the amino acid sequence of a peptide or protein may optionally be deleted providing for truncated sequences. Certain amino acids (e.g., C-terminal or N-terminal residues) may alternatively be deleted depending on the use of the sequence, as for example, expression of the sequence as part of a larger sequence which is soluble, or linked to a solid support.

“Substitutional variants” when referring to polypeptides are those that have at least one amino acid residue in a native or starting sequence removed and a different amino acid inserted in its place at the same position. The substitutions may be single, where only one amino acid in the molecule has been substituted, or they may be multiple, where two or more amino acids have been substituted in the same molecule.

As used herein the term “conservative amino acid substitution” refers to the substitution of an amino acid that is normally present in the sequence with a different amino acid of similar size, charge, or polarity. Examples of conservative substitutions include the substitution of a non-polar (hydrophobic) residue such as isoleucine, valine and leucine for another non-polar residue. Likewise, examples of conservative substitutions include the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, and between glycine and serine. Additionally, the substitution of a basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue such as aspartic acid or glutamic acid for another acidic residue are additional examples of conservative substitutions. Examples of non-conservative substitutions include the substitution of a non-polar (hydrophobic) amino acid residue such as isoleucine, valine, leucine, alanine, methionine for a polar (hydrophilic) residue such as cysteine, glutamine, glutamic acid or lysine and/or a polar residue for a non-polar residue.

“Insertional variants” when referring to polypeptides are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a native or starting sequence. “Immediately adjacent” to an amino acid means connected to either the alpha-carboxy or alpha-amino functional group of the amino acid.

“Deletional variants” when referring to polypeptides are those with one or more amino acids in the native or starting amino acid sequence removed. Ordinarily, deletional variants will have one or more amino acids deleted in a particular region of the molecule.

“Covalent derivatives” when referring to polypeptides include modifications of a native or starting protein with an organic proteinaceous or non-proteinaceous derivatizing agent, and/or post-translational modifications. Covalent modifications are traditionally introduced by reacting targeted amino acid residues of the protein with an organic derivatizing agent that is capable of reacting with selected side-chains or terminal residues, or by harnessing mechanisms of post-translational modifications that function in selected recombinant host cells. The resultant covalent derivatives are useful in programs directed at identifying residues important for biological activity, for immunoassays, or for the preparation of anti-protein antibodies for immunoaffinity purification of the recombinant glycoprotein. Such modifications are within the ordinary skill in the art and are performed without undue experimentation.

Certain post-translational modifications are the result of the action of recombinant host cells on the expressed oncology-related polypeptide. Glutaminyl and asparaginyl residues are frequently post-translationally deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues may be present in the oncology-related polypeptides produced in accordance with the present invention.

Other post-translational modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the alpha-amino groups of lysine, arginine, and histidine side chains (T. E. Creighton, Proteins: Structure and Molecular Properties, W.H. Freeman & Co., San Francisco, pp. 79-86 (1983)).

“Features” when referring to polypeptides are defined as distinct amino acid sequence-based components of a molecule. Features of the polypeptides encoded by the mmRNA of the present invention include surface manifestations, local conformational shape, folds, loops, half-loops, domains, half-domains, sites, termini or any combination thereof.

As used herein when referring to polypeptides the term “surface manifestation” refers to a polypeptide based component of a protein appearing on an outermost surface.

As used herein when referring to polypeptides the term “local conformational shape” means a polypeptide based structural manifestation of a protein which is located within a definable space of the protein.

As used herein when referring to polypeptides the term “fold” refers to the resultant conformation of an amino acid sequence upon energy minimization. A fold may occur at the secondary or tertiary level of the folding process. Examples of secondary level folds include beta sheets and alpha helices. Examples of tertiary folds include domains and regions formed due to aggregation or separation of energetic forces. Regions formed in this way include hydrophobic and hydrophilic pockets, and the like.

As used herein the term “turn” as it relates to protein conformation means a bend which alters the direction of the backbone of a peptide or polypeptide and may involve one, two, three or more amino acid residues.

As used herein when referring to polypeptides the term “loop” refers to a structural feature of a polypeptide which may serve to reverse the direction of the backbone of a peptide or polypeptide. Where the loop is found in a polypeptide and only alters the direction of the backbone, it may comprise four or more amino acid residues. Oliva et al. have identified at least 5 classes of protein loops (J. Mol Biol 266 (4): 814-830; 1997). Loops may be open or closed. Closed loops or “cyclic” loops may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids between the bridging moieties. Such bridging moieties may comprise a cysteine-cysteine bridge (Cys-Cys) typical in polypeptides having disulfide bridges or alternatively bridging moieties may be non-protein based such as the dibromozylyl agents used herein.

As used herein when referring to polypeptides the term “half-loop” refers to a portion of an identified loop having at least half the number of amino acid resides as the loop from which it is derived. It is understood that loops may not always contain an even number of amino acid residues. Therefore, in those cases where a loop contains or is identified to comprise an odd number of amino acids, a half-loop of the odd-numbered loop will comprise the whole number portion or next whole number portion of the loop (number of amino acids of the loop/2+/−0.5 amino acids). For example, a loop identified as a 7 amino acid loop could produce half-loops of 3 amino acids or 4 amino acids (7/2=3.5+/−0.5 being 3 or 4).

As used herein when referring to polypeptides the term “domain” refers to a motif of a polypeptide having one or more identifiable structural or functional characteristics or properties (e.g., binding capacity, serving as a site for protein-protein interactions).

As used herein when referring to polypeptides the term “half-domain” means a portion of an identified domain having at least half the number of amino acid resides as the domain from which it is derived. It is understood that domains may not always contain an even number of amino acid residues. Therefore, in those cases where a domain contains or is identified to comprise an odd number of amino acids, a half-domain of the odd-numbered domain will comprise the whole number portion or next whole number portion of the domain (number of amino acids of the domain/2+/−0.5 amino acids). For example, a domain identified as a 7 amino acid domain could produce half-domains of 3 amino acids or 4 amino acids (7/2=3.5+/−0.5 being 3 or 4). It is also understood that subdomains may be identified within domains or half-domains, these subdomains possessing less than all of the structural or functional properties identified in the domains or half domains from which they were derived. It is also understood that the amino acids that comprise any of the domain types herein need not be contiguous along the backbone of the polypeptide (i.e., nonadjacent amino acids may fold structurally to produce a domain, half-domain or subdomain).

As used herein when referring to polypeptides the terms “site” as it pertains to amino acid based embodiments is used synonymously with “amino acid residue” and “amino acid side chain.” A site represents a position within a peptide or polypeptide that may be modified, manipulated, altered, derivatized or varied within the polypeptide based molecules of the present invention.

As used herein the terms “termini” or “terminus” when referring to polypeptides refers to an extremity of a peptide or polypeptide. Such extremity is not limited only to the first or final site of the peptide or polypeptide but may include additional amino acids in the terminal regions. The polypeptide based molecules of the present invention may be characterized as having both an N-terminus (terminated by an amino acid with a free amino group (NH2)) and a C-terminus (terminated by an amino acid with a free carboxyl group (COOH)). Proteins of the invention are in some cases made up of multiple polypeptide chains brought together by disulfide bonds or by non-covalent forces (multimers, oligomers). These sorts of proteins will have multiple N- and C-termini. Alternatively, the termini of the polypeptides may be modified such that they begin or end, as the case may be, with a non-polypeptide based moiety such as an organic conjugate.

Once any of the features have been identified or defined as a desired component of a polypeptide to be encoded by the signal-sensor primary construct or mmRNA of the invention, any of several manipulations and/or modifications of these features may be performed by moving, swapping, inverting, deleting, randomizing or duplicating. Furthermore, it is understood that manipulation of features may result in the same outcome as a modification to the molecules of the invention. For example, a manipulation which involved deleting a domain would result in the alteration of the length of a molecule just as modification of a nucleic acid to encode less than a full length molecule would.

Modifications and manipulations can be accomplished by methods known in the art such as, but not limited to, site directed mutagenesis. The resulting modified molecules may then be tested for activity using in vitro or in vivo assays such as those described herein or any other suitable screening assay known in the art.

According to the present invention, the oncology-related polypeptides may comprise a consensus sequence which is discovered through rounds of experimentation. As used herein a “consensus” sequence is a single sequence which represents a collective population of sequences allowing for variability at one or more sites.

As recognized by those skilled in the art, protein fragments, functional protein domains, and hom*ologous proteins are also considered to be within the scope of oncology-related polypeptides of interest of this invention. For example, provided herein is any protein fragment (meaning an oncology-related polypeptide sequence at least one amino acid residue shorter than a reference oncology-related polypeptide sequence but otherwise identical) of a reference oncology-related protein 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 amino acids in length. In another example, any oncology-related protein that includes a stretch of about 20, about 30, about 40, about 50, or about 100 amino acids which are about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 100% identical to any of the sequences described herein can be utilized in accordance with the invention. In certain embodiments, a polypeptide to be utilized in accordance with the invention includes 2, 3, 4, 5, 6, 7, 8, 9, 10, or more mutations as shown in any of the sequences provided or referenced herein.

Encoded Oncology-Related Polypeptides

The signal-sensor primary constructs or mmRNA of the present invention may be designed to encode oncology-related polypeptides of interest such as oncology-related peptides and proteins.

In one embodiment, signal-sensor primary constructs or mmRNA of the present invention may encode variant polypeptides which have a certain identity with a reference oncology-related polypeptide sequence. As used herein, a “reference oncology-related polypeptide sequence” refers to a starting oncology-related polypeptide sequence. Reference sequences may be wild type sequences or any sequence to which reference is made in the design of another sequence. A “reference polypeptide sequence” may, e.g., be any one of the protein sequence listed in Table 6.

The term “identity” as known in the art, refers to a relationship between the sequences of two or more peptides, as determined by comparing the sequences. In the art, identity also means the degree of sequence relatedness between peptides, as determined by the number of matches between strings of two or more amino acid residues. Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., “algorithms”). Identity of related peptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991; and Carillo et al., SIAM J. Applied Math. 48, 1073 (1988).

In some embodiments, the polypeptide variant may have the same or a similar activity as the reference oncology-related polypeptide. Alternatively, the variant may have an altered activity (e.g., increased or decreased) relative to a reference oncology-related polypeptide. Generally, variants of a particular signal-sensor polynucleotide or oncology-related polypeptide of the invention will have at least about 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% but less than 100% sequence identity to that particular reference signal-sensor polynucleotide or oncology-related polypeptide as determined by sequence alignment programs and parameters described herein and known to those skilled in the art. Such tools for alignment include those of the BLAST suite (Stephen F. Altschul, Thomas L. Madden, Alejandro A. Schaffer, Jinghui Zhang, Zheng Zhang, Webb Miller, and David J. Lipman (1997), “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res. 25:3389-3402.) Other tools are described herein, specifically in the definition of “identity.”

Default parameters in the BLAST algorithm include, for example, an expect threshold of 10, Word size of 28, Match/Mismatch Scores 1, −2, Gap costs Linear. Any filter can be applied as well as a selection for species specific repeats, e.g., hom*o sapiens.

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be used to treat a disease, disorder and/or condition in a subject.

In one embodiment, the polynucleotides, primary constructs and/or mmRNA may be used to reduce, eliminate or prevent tumor growth in a subject.

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be used to recude and/or ameliorate at least one symptom of cancer in a subject. A symptom of cancer may include, but is not limited to, weakness, aches and pains, fever, fatigue, weight loss, blood clots, increased blood calcium levels, low white blood cell count, short of breath, dizziness, headaches, hyperpigmentation, jaundice, erthema, pruritis, excessive hair growth, change in bowel habits, change in bladder function, long-lasting sores, white patches inside the mouth, white spots on the tongue, unusual bleeding or discharge, thickening or lump on parts of the body, indigestion, trouble swallowing, changes in warts or moles, change in new skin and nagging cough or hoarseness. Further, the signal-sensor polynucleotides, primary constructs and/or mmRNA may reduce a side-effect associated with cancer such as, but not limited to, chemo brain, peripheral neuropathy, fatigue, depression, nausea, vomiting, pain, anemia, lymphedema, infections, sexual side effects, reduced fertility or infertility, ostomies, insomnia and hair loss.

Oncology-Related Proteins or Oncology-Related Peptides

The signal-sensor primary constructs or mmRNA disclosed herein, may encode one or more validated or “in testing” oncology-related proteins or oncology-related peptides.

According to the present invention, one or more oncology-related proteins or oncology-related peptides currently being marketed or in development may be encoded by the oncology-related signal-sensor polynucleotide, primary constructs or mmRNA of the present invention. While not wishing to be bound by theory, it is believed that incorporation into the signal-sensor primary constructs or mmRNA of the invention will result in improved therapeutic efficacy due at least in part to the specificity, purity and selectivity of the construct designs.

The signal-sensor polynucleotides, primary constructs and/or mmRNA may alter a biological and/or physiological process and/or compound such as, but not limited to, the cell cycle, the DNA damage response (e.g., DNA damage repair), apoptosis, angiogenesis, cell motility, the epithelial to mesenchymal transition in epithelial cells, the phosphatidyl inositol 3 (PI3) kinase/Akt cellular signaling pathway, telomerase activity and/or expression, tumor metastasis, tumorigenesis, cathepsins, cell senescence, receptor tyrosine kinase signaling, metabolism and drug metabolism, G protein signaling, growth factors and receptors, heat shock proteins, histone deacetylases, hormone receptors, hypoxia, poly ADP-ribose polymerases, protein kinases, RAS signaling, topoisomerases, transcription factors and tumor suppressor activity in cancerous, precancerous and/or other cells.

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may used to express a polypeptide in cells or tissues for the purpose of replacing the protein produced from a deleted or mutated gene.

Further, the polynucleotides, primary constructs or mmRNA of the invention may be used to treat cancer which has been caused by carcinogens of natural and/or synthetic origin. In another embodiment, the use of the polynucleotides, primary constructs and/or mmRNA may be used to treat cancer caused by other organisms and/or cancers caused by viral infection.

Sensors in the Flanking Regions: Untranslated Regions (UTRs)

Untranslated regions (UTRs) of a gene are transcribed but not translated. The 5′UTR starts at the transcription start site and continues to the start codon but does not include the start codon; whereas, the 3′UTR starts immediately following the stop codon and continues until the transcriptional termination signal. There is growing body of evidence about the regulatory roles played by the UTRs in terms of stability of the nucleic acid molecule and translation. The regulatory features of a UTR can be incorporated into the signal-sensor polynucleotides, primary constructs and/or mmRNA of the present invention to enhance the stability of the molecule. The specific features can also be incorporated to ensure controlled down-regulation of the transcript in case they are misdirected to undesired organs sites. The untranslated regions may be incorporated into a vector system which can produce mRNA and/or be delivered to a cell, tissue and/or organism to produce a polypeptide of interest.

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA of the present may comprise at least one terminal modification. Non-limiting examples of terminal modifications are described in U.S. Provisional Patent Application No. 61/729,933, filed Nov. 26, 2012, entitled Terminally Optimized Modified RNAs, U.S. Provisional Patent application No. 61/737,224, filed Dec. 14, 2012, entitled Terminally Optimized RNAs, U.S. Provisional Patent Application No. 61/758,921, filed Jan. 31, 2013, entitled Differential Targeting Using RNA Constructs, U.S. Provisional Patent Application No. 61/781,139, filed Mar. 14, 2013, entitled Differential Targeting Using RNA Constructs, U.S. Provisional Patent Application No. 61/829,359, filed May 31, 2013, entitled Differential Targeting Using RNA Constructs, U.S. Provisional Patent Application No. 61/839,903, filed Jun. 27, 2013, entitled Differential Targeting Using RNA Constructs, U.S. Provisional Patent Application No. 61/842,709, filed Jul. 3, 2013, entitled Differential Targeting Using RNA Constructs, and U.S. Provisional Patent Application No. 61/857,436, filed Jul. 23, 2013, entitled Differential Targeting Using RNA Constructs, the contents of each of which are herein incorporated by reference in their entireties. These terminal modifications include, but are not limited to, 5′ caps, microRNA binding sites in the terminal region, chain terminating nucleosides, translation enhancer elements in the terminal region and tailing sequences including a polyA-G quartet and stem loop sequences.

5′ UTR and Translation Initiation

Natural 5′UTRs bear features which play roles in for translation initiation. They harbor signatures like Kozak sequences which are commonly known to be involved in the process by which the ribosome initiates translation of many genes. Kozak sequences have the consensus CCR(A/G)CCAUGG, where R is a purine (adenine or guanine) three bases upstream of the start codon (AUG), which is followed by another ‘G’. 5′UTR also have been known to form secondary structures which are involved in elongation factor binding. For example, one of the secondary 5′-UTR structures is the structured IRES for eIF4A2 elongation factor binding, which is necessary for the microRNA mediated gene repression at 3′-UTR.

5′UTR secondary structures involved in elongation factor binding can interact with other RNA binding molecules in the 5′UTR or 3′UTR to regulate gene expression. For example, the elongation factor EIF4A2 binding to a secondarily structured element in the 5′UTR is necessary for microRNA mediated repression (Meijer H A et al., Science, 2013, 340, 82-85, herein incorporated by reference in its entirety). The different secondary structures in the 5′UTR can be incorporated into the flanking region to either stabilize or selectively destabilized mRNAs in specific tissues or cells.

By engineering the features typically found in abundantly expressed genes of specific target organs, one can enhance the stability and oncology-related protein production of the signal-sensor polynucleotides, primary constructs or mmRNA of the invention. For example, introduction of 5′ UTR of liver-expressed mRNA, such as albumin, serum amyloid A, Apolipoprotein AB/E, transferrin, alpha fetoprotein, erythropoietin, or Factor VIII, could be used to enhance expression of a nucleic acid molecule, such as a mmRNA, in hepatic cell lines or liver. Likewise, use of 5′ UTR from other tissue-specific mRNA to improve expression in that tissue is possible—for muscle (MyoD, Myosin, Myoglobin, Myogenin, Herculin), for endothelial cells (Tie-1, CD36), for myeloid cells (C/EBP, AML1, G-CSF, GM-CSF, CD11b, MSR, Fr-1, i-NOS), for leukocytes (CD45, CD18), for adipose tissue (CD36, GLUT4, ACRP30, adiponectin) and for lung epithelial cells (SP-A/B/C/D).

Other non-UTR sequences may be incorporated into the 5′ (or 3′ UTR) UTRs. For example, introns or portions of introns sequences may be incorporated into the flanking regions of the signal-sensor polynucleotides, primary constructs or mmRNA of the invention. Incorporation of intronic sequences may increase protein production as well as mRNA levels.

Translation Enhancer Elements (TEEs)

In one embodiment, the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA may include at least one translational enhancer polynucleotide, translation enhancer element, translational enhancer elements (collectively referred to as “TEE”s). As a non-limiting example, the TEE may be located between the transcription promoter and the start codon. The signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA with at least one TEE in the 5′UTR may include a cap at the 5′UTR. Further, at least one TEE may be located in the 5′UTR of signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA undergoing cap-dependent or cap-independent translation.

The term “translational enhancer element” or “translation enhancer element” (herein collectively referred to as “TEE”) refers to sequences that increase the amount of polypeptide or protein produced from an mRNA.

In one embodiment, TEEs are conserved elements in the UTR which can promote translational activity of a nucleic acid such as, but not limited to, cap-dependent or cap-independent translation. The conservation of these sequences has been previously shown by Panek et al (Nucleic Acids Research, 2013, 1-10; herein incorporated by reference in its entirety) across 14 species including humans.

In one embodiment, the TEE may be any of the TEEs listed in Table 35 in Example 45, including portion and/or fragments thereof. The TEE sequence may include at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99% or more than 99% of the TEE sequences disclosed in Table 35 and/or the TEE sequence may include a 5-30 nucleotide fragment, a 5-25 nucleotide fragment, a 5-20 nucleotide fragment, a 5-15 nucleotide fragment, a 5-10 nucleotide fragment of the TEE sequences disclosed in Table 35.

In one non-limiting example, the TEEs known may be in the 5′-leader of the Gtx homeodomain protein (Chappell et al., Proc. Natl. Acad. Sci. USA 101:9590-9594, 2004, herein incorporated by reference in their entirety).

In another non-limiting example, TEEs are disclosed as SEQ ID NOs: 1-35 in US Patent Publication No. US20090226470, SEQ ID NOs: 1-35 in US Patent Publication US20130177581, SEQ ID NOs: 1-35 in International Patent Publication No. WO2009075886, SEQ ID NOs: 1-5, and 7-645 in International Patent Publication No. WO2012009644, SEQ ID NO: 1 in International Patent Publication No. WO1999024595, SEQ ID NO: 1 in U.S. Pat. No. 6,310,197, and SEQ ID NO: 1 in U.S. Pat. No. 6,849,405, each of which is herein incorporated by reference in its entirety.

In yet another non-limiting example, the TEE may be an internal ribosome entry site (IRES), HCV-IRES or an IRES element such as, but not limited to, those described in U.S. Pat. No. 7,468,275, US Patent Publication Nos. US20070048776 and US20110124100 and International Patent Publication Nos. WO2007025008 and WO2001055369, each of which is herein incorporated by reference in its entirety. The IRES elements may include, but are not limited to, the Gtx sequences (e.g., Gtx9-nt, Gtx8-nt, Gtx7-nt) described by Chappell et al. (Proc. Natl. Acad. Sci. USA 101:9590-9594, 2004) and Zhou et al. (PNAS 102:6273-6278, 2005) and in US Patent Publication Nos. US20070048776 and US20110124100 and International Patent Publication No. WO2007025008, each of which is herein incorporated by reference in its entirety.

“Translational enhancer polynucleotides” or “translation enhancer polynucleotide sequences” are polynucleotides which include one or more of the specific TEE exemplified herein and/or disclosed in the art (see e.g., U.S. Pat. Nos. 6,310,197, 6,849,405, 7,456,273, 7,183,395, US20090226470, US20070048776, US20110124100, US20090093049, US20130177581, WO2009075886, WO2007025008, WO2012009644, WO2001055371 WO1999024595, and EP2610341A1 and EP2610340A1; each of which is herein incorporated by reference in its entirety) or their variants, hom*ologs or functional derivatives. One or multiple copies of a specific TEE can be present in the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA. The TEEs in the translational enhancer polynucleotides can be organized in one or more sequence segments. A sequence segment can harbor one or more of the specific TEEs exemplified herein, with each TEE being present in one or more copies. When multiple sequence segments are present in a translational enhancer polynucleotide, they can be hom*ogenous or heterogeneous. Thus, the multiple sequence segments in a translational enhancer polynucleotide can harbor identical or different types of the specific TEEs exemplified herein, identical or different number of copies of each of the specific TEEs, and/or identical or different organization of the TEEs within each sequence segment.

In one embodiment, the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA may include at least one TEE that is described in International Patent Publication No. WO1999024595, WO2012009644, WO2009075886, WO2007025008, WO1999024595, European Patent Publication No. EP2610341A1 and EP2610340A1, U.S. Pat. Nos. 6,310,197, 6,849,405, 7,456,273, 7,183,395, US Patent Publication No. US20090226470, US20110124100, US20070048776, US20090093049 and US20130177581, each of which is herein incorporated by reference in its entirety. The TEE may be located in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA.

In another embodiment, the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA may include at least one TEE that has at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or at least 99% identity with the TEEs described in US Patent Publication Nos. US20090226470, US20070048776, US20130177581 and US20110124100, International Patent Publication No. WO1999024595, WO2012009644, WO2009075886 and WO2007025008, European Patent Publication No. EP2610341A1 and EP2610340A1, U.S. Pat. Nos. 6,310,197, 6,849,405, 7,456,273, 7,183,395, each of which is herein incorporated by reference in its entirety.

In one embodiment, the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA may include at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18 at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55 or more than 60 TEE sequences. The TEE sequences in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may be the same or different TEE sequences. The TEE sequences may be in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than three times. In these patterns, each letter, A, B, or C represent a different TEE sequence at the nucleotide level.

In one embodiment, the 5′UTR may include a spacer to separate two TEE sequences. As a non-limiting example, the spacer may be a 15 nucleotide spacer and/or other spacers known in the art. As another non-limiting example, the 5′UTR may include a TEE sequence-spacer module repeated at least once, at least twice, at least 3 times, at least 4 times, at least 5 times, at least 6 times, at least 7 times, at least 8 times and at least 9 times or more than 9 times in the 5′UTR.

In another embodiment, the spacer separating two TEE sequences may include other sequences known in the art which may regulate the translation of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention such as, but not limited to, miR sequences described herein (e.g., miR binding sites and miR seeds). As a non-limiting example, each spacer used to separate two TEE sequences may include a different miR sequence or component of a miR sequence (e.g., miR seed sequence).

In one embodiment, the TEE in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may include at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99% or more than 99% of the TEE sequences disclosed in US Patent Publication Nos. US20090226470, US20070048776, US20130177581 and US20110124100, International Patent Publication No. WO1999024595, WO2012009644, WO2009075886 and WO2007025008, European Patent Publication No. EP2610341A1 and EP2610340A1, U.S. Pat. Nos. 6,310,197, 6,849,405, 7,456,273, 7,183,395. In another embodiment, the TEE in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may include a 5-30 nucleotide fragment, a 5-25 nucleotide fragment, a 5-20 nucleotide fragment, a 5-15 nucleotide fragment, a 5-10 nucleotide fragment of the TEE sequences disclosed in US Patent Publication Nos. US20090226470, US20070048776, US20130177581 and US20110124100, International Patent Publication No. WO1999024595, WO2012009644, WO2009075886 and WO2007025008, European Patent Publication No. EP2610341A1 and EP2610340A1, U.S. Pat. Nos. 6,310,197, 6,849,405, 7,456,273, 7,183,395; each of which are herein incorporated by reference in their entirety.

In one embodiment, the TEE in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may include at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99% or more than 99% of the TEE sequences disclosed in Chappell et al. (Proc. Natl. Acad. Sci. USA 101:9590-9594, 2004) and Thou et al. (PNAS 102:6273-6278, 2005), in Supplemental Table 1 and in Supplemental Table 2 disclosed by Wellensiek et al (Genome-wide profiling of human cap-independent translation-enhancing elements, Nature Methods, 2013; DOI:10.1038/NMETH.2522); each of which is herein incorporated by reference in its entirety. In another embodiment, the TEE in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may include a 5-30 nucleotide fragment, a 5-25 nucleotide fragment, a 5-20 nucleotide fragment, a 5-15 nucleotide fragment, a 5-10 nucleotide fragment of the TEE sequences disclosed in Chappell et al. (Proc. Natl. Acad. Sci. USA 101:9590-9594, 2004) and Zhou et al. (PNAS 102:6273-6278, 2005), in Supplemental Table 1 and in Supplemental Table 2 disclosed by Wellensiek et al (Genome-wide profiling of human cap-independent translation-enhancing elements, Nature Methods, 2013; DOI:10.1038/NMETH.2522); each of which is herein incorporated by reference in its entirety.

In one embodiment, the TEE used in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention is an IRES sequence such as, but not limited to, those described in U.S. Pat. No. 7,468,275 and International Patent Publication No. WO2001055369, each of which is herein incorporated by reference in its entirety.

In one embodiment, the TEEs used in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may be identified by the methods described in US Patent Publication No. US20070048776 and US20110124100 and International Patent Publication Nos. WO2007025008 and WO2012009644, each of which is herein incorporated by reference in its entirety.

In another embodiment, the TEEs used in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may be a transcription regulatory element described in U.S. Pat. Nos. 7,456,273 and 7,183,395, US Patent Publication No. US20090093049, and International Publication No. WO2001055371, each of which is herein incorporated by reference in their entirety. The transcription regulatory elements may be identified by methods known in the art, such as, but not limited to, the methods described in U.S. Pat. Nos. 7,456,273 and 7,183,395, US Patent Publication No. US20090093049, and International Publication No. WO2001055371, each of which is herein incorporated by reference in their entirety.

In yet another embodiment, the TEE used in the 5′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention is an oligonucleotide or portion thereof as described in U.S. Pat. Nos. 7,456,273 and 7,183,395, US Patent Publication No. US20090093049, and International Publication No. WO2001055371, each of which is herein incorporated by reference in their entirety.

The 5′ UTR comprising at least one TEE described herein may be incorporated in a monocistronic sequence such as, but not limited to, a vector system or a nucleic acid vector. As a non-limiting example, the vector systems and nucleic acid vectors may include those described in U.S. Pat. Nos. 7,456,273 and 7,183,395, US Patent Publication No. US20070048776, US20090093049 and US20110124100 and International Patent Publication Nos. WO2007025008 and WO2001055371, each of which is herein incorporated by reference in its entirety.

In one embodiment, the TEEs described herein may be located in the 5′UTR and/or the 3′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA. The TEEs located in the 3′UTR may be the same and/or different than the TEEs located in and/or described for incorporation in the 5′UTR.

In one embodiment, the 3′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA may include at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18 at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55 or more than 60 TEE sequences. The TEE sequences in the 3′UTR of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention may be the same or different TEE sequences. The TEE sequences may be in a pattern such as ABABAB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than three times. In these patterns, each letter, A, B, or C represent a different TEE sequence at the nucleotide level.

In one embodiment, the 3′UTR may include a spacer to separate two TEE sequences. As a non-limiting example, the spacer may be a 15 nucleotide spacer and/or other spacers known in the art. As another non-limiting example, the 3′UTR may include a TEE sequence-spacer module repeated at least once, at least twice, at least 3 times, at least 4 times, at least 5 times, at least 6 times, at least 7 times, at least 8 times and at least 9 times or more than 9 times in the 3′UTR.

In another embodiment, the spacer separating two TEE sequences may include other sequences known in the art which may regulate the translation of the signal-sensor polynucleotides, primary constructs, modified nucleic acids and/or mmRNA of the present invention such as, but not limited to, miR sequences described herein (e.g., miR binding sites and miR seeds). As a non-limiting example, each spacer used to separate two TEE sequences may include a different miR sequence or component of a miR sequence (e.g., miR seed sequence).

In one embodiment, the incorporation of a miR sequence and/or a TEE sequence changes the shape of the stem loop region which may increase and/or decrease translation. (see e.g, Kedde et al. A Pumilio-induced RNA structure switch in p27-3′UTR controls miR-221 and miR-22 accessibility. Nature Cell Biology. 2010, herein incorporated by reference in its entirety).

In one embodiment, the 5′UTR may comprise at least one microRNA sequence. The microRNA sequence may be, but is not limited to, a 19 or 22 nucleotide sequence and/or a microRNA sequence without the seed.

In one embodiment the microRNA sequence in the 5′UTR may be used to stabilize the nucleic acid and/or mRNA described herein.

In another embodiment, a microRNA sequence in the 5′UTR may be used to decrease the accessibility of the site of translation initiation such as, but not limited to a start codon. Matsuda et al (PLoS One. 2010 11(5):e15057; herein incorporated by reference in its entirety) used antisense locked nucleic acid (LNA) oligonucleotides and exon-junction complexes (EJCs) around a start codon (−4 to +37 where the A of the AUG codons is +1) in order to decrease the accessibility to the first start codon (AUG). Matsuda showed that altering the sequence around the start codon with an LNA or EJC the efficiency, length and structural stability of the nucleic acid or mRNA is affected. The signal-sensor polynucleotides of the present invention may comprise a microRNA sequence, instead of the LNA or EJC sequence described by Matsuda et al, near the site of translation initiation in order to decrease the accessibility to the site of translation initiation. The site of translation initiation may be prior to, after or within the microRNA sequence. As a non-limiting example, the site of translation initiation may be located within a microRNA sequence such as a seed sequence or binding site. As another non-limiting example, the site of translation initiation may be located within a miR-122 sequence such as the seed sequence or the mir-122 binding site.

In one embodiment, the nucleic acids or mRNA of the present invention comprises at least one microRNA sequence in a region of the nucleic acid or mRNA which may interact with a RNA binding protein.

RNA Motifs for RNA Binding Proteins (RBPs)

RNA binding proteins (RBPs) can regulate numerous aspects of co- and post-transcription gene expression such as, but not limited to, RNA splicing, localization, translation, turnover, polyadenylation, capping, modification, export and localization. RNA-binding domains (RBDs), such as, but not limited to, RNA recognition motif (RR) and hnRNP K-hom*ology (KH) domains, typically regulate the sequence association between RBPs and their RNA targets (Ray et al. Nature 2013. 499:172-177; herein incorporated by reference in its entirety). In one embodiment, the canonical RBDs can bind short RNA sequences. In another embodiment, the canonical RBDs can recognize structure RNAs.

In one embodiment, the nucleic acids and/or mRNA may comprise at least one RNA-binding motif such as, but not limited to a RNA-binding domain (RBD).

In one embodiment, the RBD may be any of the RBDs, fragments or variants thereof descried by Ray et al. (Nature 2013. 499:172-177; herein incorporated by reference in its entirety).

In one embodiment, the nucleic acids or mRNA of the present invention may comprise a sequence for at least one RNA-binding domain (RBDs). When the nucleic acids or mRNA of the present invention comprise more than one RBD, the RBDs do not need to be from the same species or even the same structural class.

In one embodiment, at least one flanking region (e.g., the 5′UTR and/or the 3′UTR) may comprise at least one RBD. In another embodiment, the first flanking region and the second flanking region may both comprise at least one RBD. The RBD may be the same or each of the RBDs may have at least 60% sequence identity to the other RBD. As a non-limiting example, at least on RBD may be located before, after and/or within the 3′UTR of the nucleic acid or mRNA of the present invention. As another non-limiting example, at least one RBD may be located before or within the first 300 nucleosides of the 3′UTR.

In another embodiment, the nucleic acids and/or mRNA of the present invention may comprise at least one RBD in the first region of linked nucleosides. The RBD may be located before, after or within a coding region (e.g., the ORF).

In yet another embodiment, the first region of linked nucleosides and/or at least one flanking region may comprise at least on RBD. As a non-limiting example, the first region of linked nucleosides may comprise a RBD related to splicing factors and at least one flanking region may comprise a RBD for stability and/or translation factors.

In one embodiment, the nucleic acids and/or mRNA of the present invention may comprise at least one RBD located in a coding and/or non-coding region of the nucleic acids and/or mRNA.

In one embodiment, at least one RBD may be incorporated into at least one flanking region to increase the stability of the nucleic acid and/or mRNA of the present invention.

In one embodiment, a microRNA sequence in a RNA binding protein motif may be used to decrease the accessibility of the site of translation initiation such as, but not limited to a start codon. The signal-sensor polynucleotides of the present invention may comprise a microRNA sequence, instead of the LNA or EJC sequence described by Matsuda et al, near the site of translation initiation in order to decrease the accessibility to the site of translation initiation. The site of translation initiation may be prior to, after or within the microRNA sequence. As a non-limiting example, the site of translation initiation may be located within a microRNA sequence such as a seed sequence or binding site. As another non-limiting example, the site of translation initiation may be located within a miR-122 sequence such as the seed sequence or the mir-122 binding site.

In another embodiment, an antisense locked nucleic acid (LNA) oligonucleotides and exon-junction complexes (EJCs) may be used in the RNA binding protein motif. The LNA and EJCs may be used around a start codon (−4 to +37 where the A of the AUG codons is +1) in order to decrease the accessibility to the first start codon (AUG).

3′ UTR and the AU Rich Elements

3′UTRs are known to have stretches of Adenosines and Uridines embedded in them. These AU rich signatures are particularly prevalent in genes with high rates of turnover. Based on their sequence features and functional properties, the AU rich elements (AREs) can be separated into three classes (Chen et al, 1995): Class I AREs contain several dispersed copies of an AUUUA motif within U-rich regions. C-Myc and MyoD contain class I AREs. Class II AREs possess two or more overlapping UUAUUUA(U/A)(U/A) nonamers. Molecules containing this type of AREs include GM-CSF and TNF-a. Class III ARES are less well defined. These U rich regions do not contain an AUUUA motif. c-Jun and Myogenin are two well-studied examples of this class. Most proteins binding to the AREs are known to destabilize the messenger, whereas members of the ELAV family, most notably HuR, have been documented to increase the stability of mRNA. HuR binds to AREs of all the three classes. Engineering the HuR specific binding sites into the 3′ UTR of nucleic acid molecules will lead to HuR binding and thus, stabilization of the message in vivo.

Introduction, removal or modification of 3′ UTR AU rich elements (AREs) can be used to modulate the stability of signal-sensor polynucleotides, primary constructs or mmRNA of the invention. When engineering specific polynucleotides, primary constructs or mmRNA, one or more copies of an ARE can be introduced to make polynucleotides, primary constructs or mmRNA of the invention less stable and thereby curtail translation and decrease production of the resultant protein. Likewise, AREs can be identified and removed or mutated to increase the intracellular stability and thus increase translation and production of the resultant protein. Transfection experiments can be conducted in relevant cell lines, using signal-sensor polynucleotides, primary constructs or mmRNA of the invention and protein production can be assayed at various time points post-transfection. For example, cells can be transfected with different ARE-engineering molecules and by using an ELISA kit to the relevant protein and assaying protein produced at 6 hr, 12 hr, 24 hr, 48 hr, and 7 days post-transfection.

3′ UTR and Triple Helices

In one embodiment, signal-sequence polynucleotides of the present invention may include a triple helix on the 3′ end of the signal-sequence polynucleotides. The 3′ end of the nucleic acids of the present invention may include a triple helix alone or in combination with a Poly-A tail.

In one embodiment, the signal-sequence polynucleotides of the present invention may comprise at least a first and a second U-rich region, a conserved stem loop region between the first and second region and an A-rich region. The first and second U-rich region and the A-rich region may associate to form a triple helix on the 3′ end of the nucleic acid. This triple helix may stabilize the nucleic acid, enhance the translational efficiency of the nucleic acid and/or protect the 3′ end from degradation. Exemplary triple helices include, but are not limited to, the triple helix sequence of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), MEN-β and polyadenylated nuclear (PAN) RNA (See Wilusz et al., Genes & Development 2012 26:2392-2407; herein incorporated by reference in its entirety). In one embodiment, the 3′ end of the modified nucleic acids, enhanced modified RNA or ribonucleic acids of the present invention comprises a first U-rich region comprising TTTTTCTTTT (SEQ ID NO: 1), a second U-rich region comprising TTTTGCTTTTT (SEQ ID NO: 2) or TTTTGCTTTT (SEQ ID NO: 3), an A-rich region comprising AAAAAGCAAAA (SEQ ID NO: 4). In another embodiment, the 3′ end of the nucleic acids of the present invention comprises a triple helix formation structure comprising a first U-rich region, a conserved region, a second U-rich region and an A-rich region.

In one embodiment, the triple helix may be formed from the cleavage of a MALAT1 sequence prior to the cloverleaf structure. While not meaning to be bound by theory, MALAT1 is a long non-coding RNA which, when cleaved, forms a triple helix and a tRNA-like cloverleaf structure. The MALAT1 transcript then localizes to nuclear speckles and the tRNA-like cloverleaf localizes to the cytoplasm (Wilusz et al. Cell 2008 135(5): 919-932; herein incorporated by reference in its entirety).

As a non-limiting example, the terminal end of the nucleic acid of the present invention comprising the MALAT1 sequence can then form a triple helix structure, after RNaseP cleavage from the cloverleaf structure, which stabilizes the nucleic acid (Peart et al. Non-mRNA 3′ end formation: how the other half lives; WIREs RNA 2013; herein incorporated by reference in its entirety).

In one embodiment, the signal-sequence polynucleotides described herein comprise a MALAT1 sequence. In another embodiment, the signal-sequence polynucleotides may be polyadenylated. In yet another embodiment, the signal-sequence polynucleotides is not polyadenylated but has an increased resistance to degradation compared to unmodified nucleic acids or mRNA.

In one embodiment, the signal-sequence polynucleotides of the present invention may comprise a MALAT1 sequence in the second flanking region (e.g., the 3′UTR). As a non-limiting example, the MALAT1 sequence may be human or mouse.

In another embodiment, the cloverleaf structure of the MALAT1 sequence may also undergo processing by RNaseZ and CCA adding enzyme to form a tRNA-like structure called mascRNA (MALAT1-associated small cytoplasmic RNA). As a non-limiting example, the mascRNA may encode a protein or a fragment thereof and/or may comprise a microRNA sequence. The mascRNA may comprise at least one chemical modification described herein.

Stem Loop

In one embodiment, the nucleic acids of the present invention may include a stem loop such as, but not limited to, a histone stem loop. The stem loop may be a nucleotide sequence that is about 25 or about 26 nucleotides in length such as, but not limited to, SEQ ID NOs: 7-17 as described in International Patent Publication No. WO2013103659, herein incorporated by reference in its entirety. The histone stem loop may be located 3′ relative to the coding region (e.g., at the 3′ terminus of the coding region). As a non-limiting example, the stem loop may be located at the 3′ end of a nucleic acid described herein.

In one embodiment, the stem loop may be located in the second terminal region. As a non-limiting example, the stem loop may be located within an untranslated region (e.g., 3′UTR) in the second terminal region.

In one embodiment, the nucleic acid such as, but not limited to mRNA, which comprises the histone stem loop may be stabilized by the addition of at least one chain terminating nucleoside. Not wishing to be bound by theory, the addition of at least one chain terminating nucleoside may slow the degradation of a nucleic acid and thus can increase the half-life of the nucleic acid.

In one embodiment, the chain terminating nucleoside may be, but is not limited to, those described in International Patent Publication No. WO2013103659, herein incorporated by reference in its entirety. In another embodiment, the chain terminating nucleosides which may be used with the present invention includes, but is not limited to, 3′-deoxyadenosine (cordycepin), 3′-deoxyuridine, 3′-deoxycytosine, 3′-deoxyguanosine, 3′-deoxythymine, 2′,3′-dideoxynucleosides, such as 2′,3′-dideoxyadenosine, 2′,3′-dideoxyuridine, 2′,3′-dideoxycytosine, 2′,3′-dideoxyguanosine, 2′,3′-dideoxythymine, a 2′-deoxynucleoside, or a —O-methylnucleoside.

In another embodiment, the nucleic acid such as, but not limited to mRNA, which comprises the histone stem loop may be stabilized by a modification to the 3′ region of the nucleic acid that can prevent and/or inhibit the addition of oligo(U) (see e.g., International Patent Publication No. WO2013103659, herein incorporated by reference in its entirety).

In yet another embodiment, the nucleic acid such as, but not limited to mRNA, which comprises the histone stem loop may be stabilized by the addition of an oligonucleotide that terminates in a 3′-deoxynucleoside, 2′,3′-dideoxynucleoside 3′-O-methylnucleosides, 3′-O-ethylnucleosides, 3′-arabinosides, and other modified nucleosides known in the art and/or described herein.

In one embodiment, the nucleic acids of the present invention may include a histone stem loop, a polyA tail sequence and/or a 5′ cap structure. The histone stem loop may be before and/or after the polyA tail sequence. The nucleic acids comprising the histone stem loop and a polyA tail sequence may include a chain terminating nucleoside described herein.

In another embodiment, the nucleic acids of the present invention may include a histone stem loop and a 5′ cap structure. The 5′ cap structure may include, but is not limited to, those described herein and/or known in the art.

In one embodiment, the conserved stem loop region may comprise a miR sequence described herein. As a non-limiting example, the stem loop region may comprise the seed sequence of a miR sequence described herein. In another non-limiting example, the stem loop region may comprise a miR-122 seed sequence.

In another embodiment, the conserved stem loop region may comprise a miR sequence described herein and may also include a TEE sequence.

In one embodiment, the incorporation of a miR sequence and/or a TEE sequence changes the shape of the stem loop region which may increase and/or decrease translation. (see e.g, Kedde et al. A Pumilio-induced RNA structure switch in p27-3′UTR controls miR-221 and miR-22 accessibility. Nature Cell Biology. 2010, herein incorporated by reference in its entirety).

5′ Capping

The 5′ cap structure of an mRNA is involved in nuclear export, increasing mRNA stability and binds the mRNA Cap Binding Protein (CBP), which is responsible for mRNA stability in the cell and translation competency through the association of CBP with poly(A) binding protein to form the mature cyclic mRNA species. The cap further assists the removal of 5′ proximal introns removal during mRNA splicing.

Endogenous mRNA molecules may be 5′-end capped generating a 5′-ppp-5′-triphosphate linkage between a terminal guanosine cap residue and the 5′-terminal transcribed sense nucleotide of the mRNA molecule. This 5′-guanylate cap may then be methylated to generate an N7-methyl-guanylate residue. The ribose sugars of the terminal and/or antiterminal transcribed nucleotides of the 5′ end of the mRNA may optionally also be 2′-O-methylated. 5′-decapping through hydrolysis and cleavage of the guanylate cap structure may target a nucleic acid molecule, such as an mRNA molecule, for degradation.

Modifications to the signal-sensor polynucleotides, primary constructs, and mmRNA of the present invention may generate a non-hydrolyzable cap structure preventing decapping and thus increasing mRNA half-life. Because cap structure hydrolysis requires cleavage of 5′-ppp-5′ phosphorodiester linkages, modified nucleotides may be used during the capping reaction. For example, a Vaccinia Capping Enzyme from New England Biolabs (Ipswich, Mass.) may be used with α-thio-guanosine nucleotides according to the manufacturer's instructions to create a phosphorothioate linkage in the 5′-ppp-5′ cap. Additional modified guanosine nucleotides may be used such as α-methyl-phosphonate and seleno-phosphate nucleotides.

Additional modifications include, but are not limited to, 2′-O-methylation of the ribose sugars of 5′-terminal and/or 5′-antiterminal nucleotides of the mRNA (as mentioned above) on the 2′-hydroxyl group of the sugar ring. Multiple distinct 5′-cap structures can be used to generate the 5′-cap of a nucleic acid molecule, such as an mRNA molecule.

Cap analogs, which herein are also referred to as synthetic cap analogs, chemical caps, chemical cap analogs, or structural or functional cap analogs, differ from natural (i.e. endogenous, wild-type or physiological) 5′-caps in their chemical structure, while retaining cap function. Cap analogs may be chemically (i.e. non-enzymatically) or enzymatically synthesized and/linked to a nucleic acid molecule.

For example, the Anti-Reverse Cap Analog (ARCA) cap contains two guanines linked by a 5′-5′-triphosphate group, wherein one guanine contains an N7 methyl group as well as a 3′-O-methyl group (i.e., N7,3′-O-dimethyl-guanosine-5′-triphosphate-5′-guanosine (m7G-3′mppp-G; which may equivalently be designated 3′ O-Me-m7G(5′)ppp(5′)G). The 3′-O atom of the other, unmodified, guanine becomes linked to the 5′-terminal nucleotide of the capped nucleic acid molecule (e.g. an mRNA or mmRNA). The N7- and 3′-O-methylated guanine provides the terminal moiety of the capped nucleic acid molecule (e.g. mRNA or mmRNA).

Another exemplary cap is mCAP, which is similar to ARCA but has a 2′-O-methyl group on guanosine (i.e., N7,2′-O-dimethyl-guanosine-5′-triphosphate-5′-guanosine, m7Gm-ppp-G).

While cap analogs allow for the concomitant capping of a nucleic acid molecule in an in vitro transcription reaction, up to 20% of transcripts remain uncapped. This, as well as the structural differences of a cap analog from an endogenous 5′-cap structures of nucleic acids produced by the endogenous, cellular transcription machinery, may lead to reduced translational competency and reduced cellular stability.

Signal-sensor polynucleotides, primary constructs and mmRNA of the invention may also be capped post-transcriptionally, using enzymes, in order to generate more authentic 5′-cap structures. As used herein, the phrase “more authentic” refers to a feature that closely mirrors or mimics, either structurally or functionally, an endogenous or wild type feature. That is, a “more authentic” feature is better representative of an endogenous, wild-type, natural or physiological cellular function and/or structure as compared to synthetic features or analogs, etc., of the prior art, or which outperforms the corresponding endogenous, wild-type, natural or physiological feature in one or more respects. Non-limiting examples of more authentic 5′ cap structures of the present invention are those which, among other things, have enhanced binding of cap binding proteins, increased half life, reduced susceptibility to 5′ endonucleases and/or reduced 5′ decapping, as compared to synthetic 5′ cap structures known in the art (or to a wild-type, natural or physiological 5′ cap structure). For example, recombinant Vaccinia Virus Capping Enzyme and recombinant 2′-O-methyltransferase enzyme can create a canonical 5′-5′-triphosphate linkage between the 5′-terminal nucleotide of an mRNA and a guanine cap nucleotide wherein the cap guanine contains an N7 methylation and the 5′-terminal nucleotide of the mRNA contains a 2′-O-methyl. Such a structure is termed the Cap1 structure. This cap results in a higher translational-competency and cellular stability and a reduced activation of cellular pro-inflammatory cytokines, as compared, e.g., to other 5′ cap analog structures known in the art. Cap structures include 7mG(5′)ppp(5′)N, pN2p (cap 0), 7mG(5′)ppp(5′)NlmpNp (cap 1), and 7mG(5′)-ppp(5′)NlmpN2mp (cap 2).

Because the signal-sensor polynucleotides, primary constructs or mmRNA may be capped post-transcriptionally, and because this process is more efficient, nearly 100% of the signal-sensor polynucleotides, primary constructs or mmRNA may be capped. This is in contrast to −80% when a cap analog is linked to an mRNA in the course of an in vitro transcription reaction.

According to the present invention, 5′ terminal caps may include endogenous caps or cap analogs. According to the present invention, a 5′ terminal cap may comprise a guanine analog. Useful guanine analogs include inosine, N1-methyl-guanosine, 2′fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA-guanosine, and 2-azido-guanosine.

Viral Sequences

Additional viral sequences such as, but not limited to, the translation enhancer sequence of the barley yellow dwarf virus (BYDV-PAV) can be engineered and inserted in the 3′ UTR of the signal-sensor polynucleotides, primary constructs or mmRNA of the invention and can stimulate the translation of the construct in vitro and in vivo.

Transfection experiments can be conducted in relevant cell lines at and protein production can be assayed by ELISA at 12 hr, 24 hr, 48 hr, 72 hr and day 7 post-transfection.

IRES Sequences

Further, provided are signal-sensor polynucleotides, primary constructs or mmRNA which may contain an internal ribosome entry site (IRES). First identified as a feature Picorna virus RNA, IRES plays an important role in initiating protein synthesis in absence of the 5′ cap structure. An IRES may act as the sole ribosome binding site, or may serve as one of multiple ribosome binding sites of an mRNA. signal-sensor polynucleotides, primary constructs or mmRNA containing more than one functional ribosome binding site may encode several oncology-related peptides or oncology-related polypeptides that are translated independently by the ribosomes (“multicistronic nucleic acid molecules”). When signal-sensor polynucleotides, primary constructs or mmRNA are provided with an IRES, further optionally provided is a second translatable region. Examples of IRES sequences that can be used according to the invention include without limitation, those from picornaviruses (e.g. FMDV), pest viruses (CFFV), polio viruses (PV), encephalomyocarditis viruses (ECMV), foot-and-mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), murine leukemia virus (MLV), simian immune deficiency viruses (SIV) or cricket paralysis viruses (CrPV).

Poly-A Tails

During RNA processing, a long chain of adenine nucleotides (poly-A tail) may be added to a polynucleotide such as an mRNA molecule in order to increase stability. Immediately after transcription, the 3′ end of the transcript may be cleaved to free a 3′ hydroxyl. Then poly-A polymerase adds a chain of adenine nucleotides to the RNA. The process, called polyadenylation, adds a poly-A tail that can be between 100 and 250 residues long.

It has been discovered that unique poly-A tail lengths provide certain advantages to the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention.

Generally, the length of a poly-A tail of the present invention is greater than 30 nucleotides in length. In another embodiment, the poly-A tail is greater than 35 nucleotides in length (e.g., at least or greater than about 35, 40, 45, 50, 55, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800, 1,900, 2,000, 2,500, and 3,000 nucleotides). In some embodiments, the signal-sensor polynucleotides, primary construct, or mmRNA includes from about 30 to about 3,000 nucleotides (e.g., from 30 to 50, from 30 to 100, from 30 to 250, from 30 to 500, from 30 to 750, from 30 to 1,000, from 30 to 1,500, from 30 to 2,000, from 30 to 2,500, from 50 to 100, from 50 to 250, from 50 to 500, from 50 to 750, from 50 to 1,000, from 50 to 1,500, from 50 to 2,000, from 50 to 2,500, from 50 to 3,000, from 100 to 500, from 100 to 750, from 100 to 1,000, from 100 to 1,500, from 100 to 2,000, from 100 to 2,500, from 100 to 3,000, from 500 to 750, from 500 to 1,000, from 500 to 1,500, from 500 to 2,000, from 500 to 2,500, from 500 to 3,000, from 1,000 to 1,500, from 1,000 to 2,000, from 1,000 to 2,500, from 1,000 to 3,000, from 1,500 to 2,000, from 1,500 to 2,500, from 1,500 to 3,000, from 2,000 to 3,000, from 2,000 to 2,500, and from 2,500 to 3,000).

In one embodiment, the poly-A tail is designed relative to the length of the overall signal-sensor polynucleotides, primary constructs or mmRNA. This design may be based on the length of the coding region, the length of a particular feature or region (such as the first or flanking regions), or based on the length of the ultimate product expressed from the polynucleotides, primary constructs or mmRNA.

In this context the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% greater in length than the signal-sensor polynucleotides, primary constructs or mmRNA or feature thereof. The poly-A tail may also be designed as a fraction of polynucleotides, primary constructs or mmRNA to which it belongs. In this context, the poly-A tail may be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the construct or the total length of the construct minus the poly-A tail.

In one embodiment, engineered binding sites and/or conjugation of signal-sensor polynucleotides, primary constructs or mmRNA for Poly-A binding protein may be used to enhance expression. The engineered binding sites may be sensor sequences which can operate as binding sites for ligands of the local microenvironment of the nucleic acids and/or mRNA. As a non-limiting example, the nucleic acids and/or mRNA may comprise at least one engineered binding site to alter the binding affinity of Poly-A binding protein (PABP) and analogs thereof. The incorporation of at least one engineered binding site may increase the binding affinity of the PABP and analogs thereof.

Additionally, multiple distinct signal-sensor polynucleotides, primary constructs or mmRNA may be linked together to the PABP (Poly-A binding protein) through the 3′-end using modified nucleotides at the 3′-terminus of the poly-A tail. Transfection experiments can be conducted in relevant cell lines and protein production can be assayed by ELISA at 12 hr, 24 hr, 48 hr, 72 hr and day 7 post-transfection. As a non-limiting example, the transfection experiments may be used to evaluate the effect on PABP or analogs thereof binding affinity as a result of the addition of at least one engineered binding site.

In one embodiment, the signal-sensor polynucleotides and primary constructs of the present invention are designed to include a polyA-G Quartet. The G-quartet is a cyclic hydrogen bonded array of four guanine nucleotides that can be formed by G-rich sequences in both DNA and RNA. In this embodiment, the G-quartet is incorporated at the end of the poly-A tail. The resultant mmRNA construct is assayed for stability, protein production and other parameters including half-life at various time points. It has been discovered that the polyA-G quartet results in protein production equivalent to at least 75% of that seen using a poly-A tail of 120 nucleotides alone.

In one embodiment, the nucleic acids or mRNA of the present invention may comprise a polyA tail and may be stabilized by the addition of a chain terminating nucleoside. The nucleic acids and/or mRNA with a polyA tail may further comprise a 5′ cap structure.

In another embodiment, the nucleic acids or mRNA of the present invention may comprise a polyA-G Quartet. The nucleic acids and/or mRNA with a polyA-G Quartet may further comprise a 5′ cap structure.

In one embodiment, the chain terminating nucleoside which may be used to stabilize the nucleic acid or mRNA comprising a polyA tail or polyA-G Quartet may be, but is not limited to, those described in International Patent Publication No. WO2013103659, herein incorporated by reference in its entirety. In another embodiment, the chain terminating nucleosides which may be used with the present invention includes, but is not limited to, 3′-deoxyadenosine (cordycepin), 3′-deoxyuridine, 3′-deoxycytosine, 3′-deoxyguanosine, 3′-deoxythymine, 2′,3′-dideoxynucleosides, such as 2′,3′-dideoxyadenosine, 2′,3′-dideoxyuridine, 2′,3′-dideoxycytosine, 2′,3′-dideoxyguanosine, 2′,3′-dideoxythymine, a 2′-deoxynucleoside, or a —O-methylnucleoside.

In another embodiment, the nucleic acid such as, but not limited to mRNA, which comprise a polyA tail or a polyA-G Quartet may be stabilized by a modification to the 3′ region of the nucleic acid that can prevent and/or inhibit the addition of oligo(U) (see e.g., International Patent Publication No. WO2013103659, herein incorporated by reference in its entirety).

In yet another embodiment, the nucleic acid such as, but not limited to mRNA, which comprise a polyA tail or a polyA-G Quartet may be stabilized by the addition of an oligonucleotide that terminates in a 3′-deoxynucleoside, 2′,3′-dideoxynucleoside 3′-0-methylnucleosides, 3′-0-ethylnucleosides, 3′-arabinosides, and other modified nucleosides known in the art and/or described herein.

Quantification

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be quantified in exosomes derived from one or more bodily fluid. As used herein “bodily fluids” include peripheral blood, serum, plasma, ascites, urine, cerebrospinal fluid (CSF), sputum, saliva, bone marrow, synovial fluid, aqueous humor, amniotic fluid, cerumen, breast milk, bronchioalveolar lavage fluid, sem*n, prostatic fluid, cowper's fluid or pre-ejacul*tory fluid, sweat, fecal matter, hair, tears, cyst fluid, pleural and peritoneal fluid, pericardial fluid, lymph, chyme, chyle, bile, interstitial fluid, menses, pus, sebum, vomit, vagin*l secretions, mucosal secretion, stool water, pancreatic juice, lavage fluids from sinus cavities, bronchopulmonary aspirates, blastocyl cavity fluid, and umbilical cord blood. Alternatively, exosomes may be retrieved from an organ selected from the group consisting of lung, heart, pancreas, stomach, intestine, bladder, kidney, ovary, testis, skin, colon, breast, prostate, brain, esophagus, liver, and placenta.

In the quantification method, a sample of not more than 2 mL is obtained from the subject and the exosomes isolated by size exclusion chromatography, density gradient centrifugation, differential centrifugation, nanomembrane ultrafiltration, immunoabsorbent capture, affinity purification, microfluidic separation, or combinations thereof. In the analysis, the level or concentration of signal-sensor polynucleotides, primary construct or mmRNA may be an expression level, presence, absence, truncation or alteration of the administered construct. It is advantageous to correlate the level with one or more clinical phenotypes or with an assay for a human disease biomarker. The assay may be performed using construct specific probes, cytometry, qRT-PCR, real-time PCR, PCR, flow cytometry, electrophoresis, mass spectrometry, or combinations thereof while the exosomes may be isolated using immunohistochemical methods such as enzyme linked immunosorbent assay (ELISA) methods. Exosomes may also be isolated by size exclusion chromatography, density gradient centrifugation, differential centrifugation, nanomembrane ultrafiltration, immunoabsorbent capture, affinity purification, microfluidic separation, or combinations thereof.

These methods afford the investigator the ability to monitor, in real time, the level of signal-sensor polynucleotides, primary constructs or mmRNA remaining or delivered. This is possible because the polynucleotides, primary constructs or mmRNA of the present invention differ from the endogenous forms due to the structural and/or chemical modifications.

Signal-sensor polynucleotides, primary constructs or mmRNA for use in accordance with the invention may be prepared according to any available technique including, but not limited to chemical synthesis, enzymatic synthesis, which is generally termed in vitro transcription (IVT) or enzymatic or chemical cleavage of a longer precursor, etc. Methods of synthesizing RNAs are known in the art (see, e.g., Gait, M. J. (ed.) Oligonucleotide synthesis: a practical approach, Oxford [Oxfordshire], Washington, D.C.: IRL Press, 1984; and Herdewijn, P. (ed.) Oligonucleotide synthesis: methods and applications, Methods in Molecular Biology, v. 288 (Clifton, N.J.) Totowa, N.J.: Humana Press, 2005; both of which are incorporated herein by reference).

The process of design and synthesis of the signal-sensor primary constructs of the invention generally includes the steps of gene construction, mRNA production (either with or without modifications) and purification. In the enzymatic synthesis method, a target signal-sensor polynucleotide sequence encoding the oncology-related polypeptide of interest is first selected for incorporation into a vector which will be amplified to produce a cDNA template. Optionally, the target signal-sensor polynucleotide sequence and/or any flanking sequences may be codon optimized. The cDNA template is then used to produce mRNA through in vitro transcription (IVT). After production, the mRNA may undergo purification and clean-up processes. The steps of which are provided in more detail below.

Gene Construction

The step of gene construction may include, but is not limited to gene synthesis, vector amplification, plasmid purification, plasmid linearization and clean-up, and cDNA template synthesis and clean-up.

Gene Synthesis

Once an oncology-related polypeptide of interest, or target, is selected for production, a signal-sensor primary construct is designed. Within the primary construct, a first region of linked nucleosides encoding the polypeptide of interest may be constructed using an open reading frame (ORF) of a selected nucleic acid (DNA or RNA) transcript. The ORF may comprise the wild type ORF, an isoform, variant or a fragment thereof. As used herein, an “open reading frame” or “ORF” is meant to refer to a nucleic acid sequence (DNA or RNA) which is capable of encoding an oncology-related polypeptide of interest. ORFs often begin with the start codon, ATG and end with a nonsense or termination codon or signal.

Further, the nucleotide sequence of the first region may be codon optimized. Codon optimization methods are known in the art and may be useful in efforts to achieve one or more of several goals. These goals include to match codon frequencies in target and host organisms to ensure proper folding, bias GC content to increase mRNA stability or reduce secondary structures, minimize tandem repeat codons or base runs that may impair gene construction or expression, customize transcriptional and translational control regions, insert or remove protein trafficking sequences, remove/add post translation modification sites in encoded protein (e.g. glycosylation sites), add, remove or shuffle protein domains, insert or delete restriction sites, modify ribosome binding sites and mRNA degradation sites, to adjust translational rates to allow the various domains of the protein to fold properly, or to reduce or eliminate problem secondary structures within the mRNA. Codon optimization tools, algorithms and services are known in the art, non-limiting examples include services from GeneArt (Life Technologies) and/or DNA2.0 (Menlo Park Calif.). In one embodiment, the ORF sequence is optimized using optimization algorithms. Codon options for each amino acid are given in Table 1.

TABLE 1
Codon Options
Single
Letter
Amino AcidCodeCodon Options
IsoleucineIATT, ATC, ATA
LeucineLCTT, CTC, CTA, CTG, TTA, TTG
ValineVGTT, GTC, GTA, GTG
PhenylalanineFTTT, TTC
MethionineMATG
CysteineCTGT, TGC
AlanineAGCT, GCC, GCA, GCG
GlycineGGGT, GGC, GGA, GGG
ProlinePCCT, CCC, CCA, CCG
ThreonineTACT, ACC, ACA, ACG
SerineSTCT, TCC, TCA, TCG, AGT, AGC
TyrosineYTAT, TAC
TryptophanWTGG
GlutamineQCAA, CAG
AsparagineNAAT, AAC
HistidineHCAT, CAC
Glutamic acidEGAA, GAG
Aspartic acidDGAT, GAC
LysineKAAA, AAG
ArginineRCGT, CGC, CGA, CGG, AGA, AGG
SelenocysteineSecUGA in mRNA in presence of Selenocystein
insertion element (SECIS)
Stop codonsStopTAA, TAG, TGA

In one embodiment, after a nucleotide sequence has been codon optimized it may be further evaluated for regions containing restriction sites. At least one nucleotide within the restriction site regions may be replaced with another nucleotide in order to remove the restriction site from the sequence but the replacement of nucleotides does alter the amino acid sequence which is encoded by the codon optimized nucleotide sequence.

Features, which may be considered beneficial in some embodiments of the present invention, may be encoded by the signal-sensor primary construct and may flank the ORF as a first or second flanking region. The flanking regions may be incorporated into the signal-sensor primary construct before and/or after optimization of the ORF. It is not required that a signal-sensor primary construct contain both a 5′ and 3′ flanking region. Examples of such features include, but are not limited to, untranslated regions (UTRs), Kozak sequences, an oligo(dT) sequence, and detectable tags and may include multiple cloning sites which may have XbaI recognition.

In some embodiments, a 5′ UTR and/or a 3′ UTR may be provided as flanking regions. Multiple 5′ or 3′ UTRs may be included in the flanking regions and may be the same or of different sequences. Any portion of the flanking regions, including none, may be codon optimized and any may independently contain one or more different structural or chemical modifications, before and/or after codon optimization. Combinations of features may be included in the first and second flanking regions and may be contained within other features. For example, the ORF may be flanked by a 5′ UTR which may contain a strong Kozak translational initiation signal and/or a 3′ UTR which may include an oligo(dT) sequence for templated addition of a poly-A tail.

Tables 2 and 3 provide a listing of exemplary UTRs which may be utilized in the signal-sensor primary construct of the present invention as flanking regions. Shown in Table 2 is a representative listing of a 5′-untranslated region of the invention. Variants of 5′ UTRs may be utilized wherein one or more nucleotides are added or removed to the termini, including A, T, C or G.

TABLE 2
5′-Untranslated Regions
5′ UTRName/SEQ ID
IdentifierDescriptionSequenceNO.
NativeWild type UTRSee wild type sequence
5UTR-001Synthetic UTRGGGAAATAAGAGAGAAAAGAAGAGTAAGA1
AGAAATATAAGAGCCACC
5UTR-002Upstream UTRGGGAGATCAGAGAGAAAAGAAGAGTAAGA2
AGAAATATAAGAGCCACC
5UTR-003Upstream UTRGGAATAAAAGTCTCAACACAACATATACAA3
AACAAACGAATCTCAAGCAATCAAGCATTC
TACTTCTATTGCAGCAATTTAAATCATTTCT
TTTAAAGCAAAAGCAATTTTCTGAAAATTT
TCACCATTTACGAACGATAGCAAC
5UTR-004Upstream UTRGGGAGACAAGCUUGGCAUUCCGGUACUGU4
UGGUAAAGCCACC

Shown in Table 3 is a representative listing of 3′-untranslated regions of the invention. Variants of 3′ UTRs may be utilized wherein one or more nucleotides are added or removed to the termini, including A, T, C or G.

TABLE 3
3′-Untranslated Regions
SEQ
3′ UTRName/ID
IdentifierDescriptionSequenceNO.
3UTR-001CreatineGCGCCTGCCCACCTGCCACCGACTGCTGGAACC5
KinaseCAGCCAGTGGGAGGGCCTGGCCCACCAGAGTCC
TGCTCCCTCACTCCTCGCCCCGCCCCCTGTCCCA
GAGTCCCACCTGGGGGCTCTCTCCACCCTTCTCA
GAGTTCCAGTTTCAACCAGAGTTCCAACCAATG
GGCTCCATCCTCTGGATTCTGGCCAATGAAATAT
CTCCCTGGCAGGGTCCTCTTCTTTTCCCAGAGCT
CCACCCCAACCAGGAGCTCTAGTTAATGGAGAG
CTCCCAGCACACTCGGAGCTTGTGCTTTGTCTCC
ACGCAAAGCGATAAATAAAAGCATTGGTGGCCT
TTGGTCTTTGAATAAAGCCTGAGTAGGAAGTCTA
GA
3UTR-002MyoglobinGCCCCTGCCGCTCCCACCCCCACCCATCTGGGCC6
CCGGGTTCAAGAGAGAGCGGGGTCTGATCTCGT
GTAGCCATATAGAGTTTGCTTCTGAGTGTCTGCT
TTGTTTAGTAGAGGTGGGCAGGAGGAGCTGAGG
GGCTGGGGCTGGGGTGTTGAAGTTGGCTTTGCAT
GCCCAGCGATGCGCCTCCCTGTGGGATGTCATCA
CCCTGGGAACCGGGAGTGGCCCTTGGCTCACTG
TGTTCTGCATGGTTTGGATCTGAATTAATTGTCC
TTTCTTCTAAATCCCAACCGAACTTCTTCCAACC
TCCAAACTGGCTGTAACCCCAAATCCAAGCCATT
AACTACACCTGACAGTAGCAATTGTCTGATTAAT
CACTGGCCCCTTGAAGACAGCAGAATGTCCCTTT
GCAATGAGGAGGAGATCTGGGCTGGGCGGGCCA
GCTGGGGAAGCATTTGACTATCTGGAACTTGTGT
GTGCCTCCTCAGGTATGGCAGTGACTCACCTGGT
TTTAATAAAACAACCTGCAACATCTCATGGTCTT
TGAATAAAGCCTGAGTAGGAAGTCTAGA
3UTR-003α-actinACACACTCCACCTCCAGCACGCGACTTCTCAGG7
ACGACGAATCTTCTCAATGGGGGGGCGGCTGAG
CTCCAGCCACCCCGCAGTCACTTTCTTTGTAACA
ACTTCCGTTGCTGCCATCGTAAACTGACACAGTG
TTTATAACGTGTACATACATTAACTTATTACCTC
ATTTTGTTATTTTTCGAAACAAAGCCCTGTGGAA
GAAAATGGAAAACTTGAAGAAGCATTAAAGTCA
TTCTGTTAAGCTGCGTAAATGGTCTTTGAATAAA
GCCTGAGTAGGAAGTCTAGA
3UTR-004AlbuminCATCACATTTAAAAGCATCTCAGCCTACCATGAG8
AATAAGAGAAAGAAAATGAAGATCAAAAGCTT
ATTCATCTGTTTTTCTTTTTCGTTGGTGTAAAGCC
AACACCCTGTCTAAAAAACATAAATTTCTTTAAT
CATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAA
AAAATGGAAAGAATCTAATAGAGTGGTACAGCA
CTGTTATTTTTCAAAGATGTGTTGCTATCCTGAA
AATTCTGTAGGTTCTGTGGAAGTTCCAGTGTTCT
CTCTTATTCCACTTCGGTAGAGGATTTCTAGTTT
CTTGTGGGCTAATTAAATAAATCATTAATACTCT
TCTAATGGTCTTTGAATAAAGCCTGAGTAGGAA
GTCTAGA
3UTR-005α-globinGCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGC9
CCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTC
TTTGAATAAAGCCTGAGTAGGAAGGCGGCCGCT
CGAGCATGCATCTAGA
3UTR-006G-CSFGCCAAGCCCTCCCCATCCCATGTATTTATCTCTA10
TTTAATATTTATGTCTATTTAAGCCTCATATTTAA
AGACAGGGAAGAGCAGAACGGAGCCCCAGGCC
TCTGTGTCCTTCCCTGCATTTCTGAGTTTCATTCT
CCTGCCTGTAGCAGTGAGAAAAAGCTCCTGTCCT
CCCATCCCCTGGACTGGGAGGTAGATAGGTAAA
TACCAAGTATTTATTACTATGACTGCTCCCCAGC
CCTGGCTCTGCAATGGGCACTGGGATGAGCCGC
TGTGAGCCCCTGGTCCTGAGGGTCCCCACCTGGG
ACCCTTGAGAGTATCAGGTCTCCCACGTGGGAG
ACAAGAAATCCCTGTTTAATATTTAAACAGCAGT
GTTCCCCATCTGGGTCCTTGCACCCCTCACTCTG
GCCTCAGCCGACTGCACAGCGGCCCCTGCATCC
CCTTGGCTGTGAGGCCCCTGGACAAGCAGAGGT
GGCCAGAGCTGGGAGGCATGGCCCTGGGGTCCC
ACGAATTTGCTGGGGAATCTCGTTTTTCTTCTTA
AGACTTTTGGGACATGGTTTGACTCCCGAACATC
ACCGACGCGTCTCCTGTTTTTCTGGGTGGCCTCG
GGACACCTGCCCTGCCCCCACGAGGGTCAGGAC
TGTGACTCTTTTTAGGGCCAGGCAGGTGCCTGGA
CATTTGCCTTGCTGGACGGGGACTGGGGATGTG
GGAGGGAGCAGACAGGAGGAATCATGTCAGGC
CTGTGTGTGAAAGGAAGCTCCACTGTCACCCTCC
ACCTCTTCACCCCCCACTCACCAGTGTCCCCTCC
ACTGTCACATTGTAACTGAACTTCAGGATAATAA
AGTGTTTGCCTCCATGGTCTTTGAATAAAGCCTG
AGTAGGAAGGCGGCCGCTCGAGCATGCATCTAGA
3UTR-007Col1a2;ACTCAATCTAAATTAAAAAAGAAAGAAATTTGA11
collagen,AAAAACTTTCTCTTTGCCATTTCTTCTTCTTCTTT
type I, alpha 2TTTAACTGAAAGCTGAATCCTTCCATTTCTTCTG
CACATCTACTTGCTTAAATTGTGGGCAAAAGAG
AAAAAGAAGGATTGATCAGAGCATTGTGCAATA
CAGTTTCATTAACTCCTTCCCCCGCTCCCCCAAA
AATTTGAATTTTTTTTTCAACACTCTTACACCTGT
TATGGAAAATGTCAACCTTTGTAAGAAAACCAA
AATAAAAATTGAAAAATAAAAACCATAAACATT
TGCACCACTTGTGGCTTTTGAATATCTTCCACAG
AGGGAAGTTTAAAACCCAAACTTCCAAAGGTTT
AAACTACCTCAAAACACTTTCCCATGAGTGTGAT
CCACATTGTTAGGTGCTGACCTAGACAGAGATG
AACTGAGGTCCTTGTTTTGTTTTGTTCATAATAC
AAAGGTGCTAATTAATAGTATTTCAGATACTTGA
AGAATGTTGATGGTGCTAGAAGAATTTGAGAAG
AAATACTCCTGTATTGAGTTGTATCGTGTGGTGT
ATTTTTTAAAAAATTTGATTTAGCATTCATATTTT
CCATCTTATTCCCAATTAAAAGTATGCAGATTAT
TTGCCCAAATCTTCTTCAGATTCAGCATTTGTTCT
TTGCCAGTCTCATTTTCATCTTCTTCCATGGTTCC
ACAGAAGCTTTGTTTCTTGGGCAAGCAGAAAAA
TTAAATTGTACCTATTTTGTATATGTGAGATGTT
TAAATAAATTGTGAAAAAAATGAAATAAAGCAT
GTTTGGTTTTCCAAAAGAACATAT
3UTR-008Col6a2;CGCCGCCGCCCGGGCCCCGCAGTCGAGGGTCGT12
collagen,GAGCCCACCCCGTCCATGGTGCTAAGCGGGCCC
type VI,GGGTCCCACACGGCCAGCACCGCTGCTCACTCG
alpha 2GACGACGCCCTGGGCCTGCACCTCTCCAGCTCCT
CCCACGGGGTCCCCGTAGCCCCGGCCCCCGCCC
AGCCCCAGGTCTCCCCAGGCCCTCCGCAGGCTG
CCCGGCCTCCCTCCCCCTGCAGCCATCCCAAGGC
TCCTGACCTACCTGGCCCCTGAGCTCTGGAGCAA
GCCCTGACCCAATAAAGGCTTTGAACCCAT
3UTR-009RPN1;GGGGCTAGAGCCCTCTCCGCACAGCGTGGAGAC13
ribophorin IGGGGCAAGGAGGGGGGTTATTAGGATTGGTGGT
TTTGTTTTGCTTTGTTTAAAGCCGTGGGAAAATG
GCACAACTTTACCTCTGTGGGAGATGCAACACT
GAGAGCCAAGGGGTGGGAGTTGGGATAATTTTT
ATATAAAAGAAGTTTTTCCACTTTGAATTGCTAA
AAGTGGCATTTTTCCTATGTGCAGTCACTCCTCT
CATTTCTAAAATAGGGACGTGGCCAGGCACGGT
GGCTCATGCCTGTAATCCCAGCACTTTGGGAGGC
CGAGGCAGGCGGCTCACGAGGTCAGGAGATCGA
GACTATCCTGGCTAACACGGTAAAACCCTGTCTC
TACTAAAAGTACAAAAAATTAGCTGGGCGTGGT
GGTGGGCACCTGTAGTCCCAGCTACTCGGGAGG
CTGAGGCAGGAGAAAGGCATGAATCCAAGAGG
CAGAGCTTGCAGTGAGCTGAGATCACGCCATTG
CACTCCAGCCTGGGCAACAGTGTTAAGACTCTGT
CTCAAATATAAATAAATAAATAAATAAATAAAT
AAATAAATAAAAATAAAGCGAGATGTTGCCCTC
AAA
3UTR-010LRP1; lowGGCCCTGCCCCGTCGGACTGCCCCCAGAAAGCC14
densityTCCTGCCCCCTGCCAGTGAAGTCCTTCAGTGAGC
lipoproteinCCCTCCCCAGCCAGCCCTTCCCTGGCCCCGCCGG
receptor-ATGTATAAATGTAAAAATGAAGGAATTACATTT
relatedTATATGTGAGCGAGCAAGCCGGCAAGCGAGCAC
protein 1AGTATTATTTCTCCATCCCCTCCCTGCCTGCTCCT
TGGCACCCCCATGCTGCCTTCAGGGAGACAGGC
AGGGAGGGCTTGGGGCTGCACCTCCTACCCTCC
CACCAGAACGCACCCCACTGGGAGAGCTGGTGG
TGCAGCCTTCCCCTCCCTGTATAAGACACTTTGC
CAAGGCTCTCCCCTCTCGCCCCATCCCTGCTTGC
CCGCTCCCACAGCTTCCTGAGGGCTAATTCTGGG
AAGGGAGAGTTCTTTGCTGCCCCTGTCTGGAAG
ACGTGGCTCTGGGTGAGGTAGGCGGGAAAGGAT
GGAGTGTTTTAGTTCTTGGGGGAGGCCACCCCA
AACCCCAGCCCCAACTCCAGGGGCACCTATGAG
ATGGCCATGCTCAACCCCCCTCCCAGACAGGCC
CTCCCTGTCTCCAGGGCCCCCACCGAGGTTCCCA
GGGCTGGAGACTTCCTCTGGTAAACATTCCTCCA
GCCTCCCCTCCCCTGGGGACGCCAAGGAGGTGG
GCCACACCCAGGAAGGGAAAGCGGGCAGCCCC
GTTTTGGGGACGTGAACGTTTTAATAATTTTTGC
TGAATTCCTTTACAACTAAATAACACAGATATTG
TTATAAATAAAATTGT
3UTR-011Nnt1;ATATTAAGGATCAAGCTGTTAGCTAATAATGCC15
cardiotrophin-ACCTCTGCAGTTTTGGGAACAGGCAAATAAAGT
likeATCAGTATACATGGTGATGTACATCTGTAGCAA
cytokineAGCTCTTGGAGAAAATGAAGACTGAAGAAAGCA
factor 1AAGCAAAAACTGTATAGAGAGATTTTTCAAAAG
CAGTAATCCCTCAATTTTAAAAAAGGATTGAAA
ATTCTAAATGTCTTTCTGTGCATATTTTTTGTGTT
AGGAATCAAAAGTATTTTATAAAAGGAGAAAGA
ACAGCCTCATTTTAGATGTAGTCCTGTTGGATTT
TTTATGCCTCCTCAGTAACCAGAAATGTTTTAAA
AAACTAAGTGTTTAGGATTTCAAGACAACATTAT
ACATGGCTCTGAAATATCTGACACAATGTAAAC
ATTGCAGGCACCTGCATTTTATGTTTTTTTTTTCA
ACAAATGTGACTAATTTGAAACTTTTATGAACTT
CTGAGCTGTCCCCTTGCAATTCAACCGCAGTTTG
AATTAATCATATCAAATCAGTTTTAATTTTTTAA
ATTGTACTTCAGAGTCTATATTTCAAGGGCACAT
TTTCTCACTACTATTTTAATACATTAAAGGACTA
AATAATCTTTCAGAGATGCTGGAAACAAATCAT
TTGCTTTATATGTTTCATTAGAATACCAATGAAA
CATACAACTTGAAAATTAGTAATAGTATTTTTGA
AGATCCCATTTCTAATTGGAGATCTCTTTAATTT
CGATCAACTTATAATGTGTAGTACTATATTAAGT
GCACTTGAGTGGAATTCAACATTTGACTAATAA
AATGAGTTCATCATGTTGGCAAGTGATGTGGCA
ATTATCTCTGGTGACAAAAGAGTAAAATCAAAT
ATTTCTGCCTGTTACAAATATCAAGGAAGACCTG
CTACTATGAAATAGATGACATTAATCTGTCTTCA
CTGTTTATAATACGGATGGATTTTTTTTCAAATC
AGTGTGTGTTTTGAGGTCTTATGTAATTGATGAC
ATTTGAGAGAAATGGTGGCTTTTTTTAGCTACCT
CTTTGTTCATTTAAGCACCAGTAAAGATCATGTC
TTTTTATAGAAGTGTAGATTTTCTTTGTGACTTTG
CTATCGTGCCTAAAGCTCTAAATATAGGTGAATG
TGTGATGAATACTCAGATTATTTGTCTCTCTATA
TAATTAGTTTGGTACTAAGTTTCTCAAAAAATTA
TTAACACATGAAAGACAATCTCTAAACCAGAAA
AAGAAGTAGTACAAATTTTGTTACTGTAATGCTC
GCGTTTAGTGAGTTTAAAACACACAGTATCTTTT
GGTTTTATAATCAGTTTCTATTTTGCTGTGCCTGA
GATTAAGATCTGTGTATGTGTGTGTGTGTGTGTG
TGCGTTTGTGTGTTAAAGCAGAAAAGACTTTTTT
AAAAGTTTTAAGTGATAAATGCAATTTGTTAATT
GATCTTAGATCACTAGTAAACTCAGGGCTGAATT
ATACCATGTATATTCTATTAGAAGAAAGTAAAC
ACCATCTTTATTCCTGCCCTTTTTCTTCTCTCAAA
GTAGTTGTAGTTATATCTAGAAAGAAGCAATTTT
GATTTCTTGAAAAGGTAGTTCCTGCACTCAGTTT
AAACTAAAAATAATCATACTTGGATTTTATTTAT
TTTTGTCATAGTAAAAATTTTAATTTATATATATT
TTTATTTAGTATTATCTTATTCTTTGCTATTTGCC
AATCCTTTGTCATCAATTGTGTTAAATGAATTGA
AAATTCATGCCCTGTTCATTTTATTTTACTTTATT
GGTTAGGATATTTAAAGGATTTTTGTATATATAA
TTTCTTAAATTAATATTCCAAAAGGTTAGTGGAC
TTAGATTATAAATTATGGCAAAAATCTAAAAAC
AACAAAAATGATTTTTATACATTCTATTTCATTA
TTCCTCTTTTTCCAATAAGTCATACAATTGGTAG
ATATGACTTATTTTATTTTTGTATTATTCACTATA
TCTTTATGATATTTAAGTATAAATAATTAAAAAA
ATTTATTGTACCTTATAGTCTGTCACCAAAAAAA
AAAAATTATCTGTAGGTAGTGAAATGCTAATGTT
GATTTGTCTTTAAGGGCTTGTTAACTATCCTTTAT
TTTCTCATTTGTCTTAAATTAGGAGTTTGTGTTTA
AATTACTCATCTAAGCAAAAAATGTATATAAAT
CCCATTACTGGGTATATACCCAAAGGATTATAA
ATCATGCTGCTATAAAGACACATGCACACGTAT
GTTTATTGCAGCACTATTCACAATAGCAAAGACT
TGGAACCAACCCAAATGTCCATCAATGATAGAC
TTGATTAAGAAAATGTGCACATATACACCATGG
AATACTATGCAGCCATAAAAAAGGATGAGTTCA
TGTCCTTTGTAGGGACATGGATAAAGCTGGAAA
CCATCATTCTGAGCAAACTATTGCAAGGACAGA
AAACCAAACACTGCATGTTCTCACTCATAGGTG
GGAATTGAACAATGAGAACACTTGGACACAAGG
TGGGGAACACCACACACCAGGGCCTGTCATGGG
GTGGGGGGAGTGGGGAGGGATAGCATTAGGAG
ATATACCTAATGTAAATGATGAGTTAATGGGTG
CAGCACACCAACATGGCACATGTATACATATGT
AGCAAACCTGCACGTTGTGCACATGTACCCTAG
AACTTAAAGTATAATTAAAAAAAAAAAGAAAAC
AGAAGCTATTTATAAAGAAGTTATTTGCTGAAAT
AAATGTGATCTTTCCCATTAAAAAAATAAAGAA
ATTTTGGGGTAAAAAAACACAATATATTGTATTC
TTGAAAAATTCTAAGAGAGTGGATGTGAAGTGT
TCTCACCACAAAAGTGATAACTAATTGAGGTAA
TGCACATATTAATTAGAAAGATTTTGTCATTCCA
CAATGTATATATACTTAAAAATATGTTATACACA
ATAAATACATACATTAAAAAATAAGTAAATGTA
3UTR-012Col6a1;CCCACCCTGCACGCCGGCACCAAACCCTGTCCTC16
collagen,CCACCCCTCCCCACTCATCACTAAACAGAGTAA
type VI,AATGTGATGCGAATTTTCCCGACCAACCTGATTC
alpha 1GCTAGATTTTTTTTAAGGAAAAGCTTGGAAAGCC
AGGACACAACGCTGCTGCCTGCTTTGTGCAGGG
TCCTCCGGGGCTCAGCCCTGAGTTGGCATCACCT
GCGCAGGGCCCTCTGGGGCTCAGCCCTGAGCTA
GTGTCACCTGCACAGGGCCCTCTGAGGCTCAGC
CCTGAGCTGGCGTCACCTGTGCAGGGCCCTCTGG
GGCTCAGCCCTGAGCTGGCCTCACCTGGGTTCCC
CACCCCGGGCTCTCCTGCCCTGCCCTCCTGCCCG
CCCTCCCTCCTGCCTGCGCAGCTCCTTCCCTAGG
CACCTCTGTGCTGCATCCCACCAGCCTGAGCAAG
ACGCCCTCTCGGGGCCTGTGCCGCACTAGCCTCC
CTCTCCTCTGTCCCCATAGCTGGTTTTTCCCACCA
ATCCTCACCTAACAGTTACTTTACAATTAAACTC
AAAGCAAGCTCTTCTCCTCAGCTTGGGGCAGCC
ATTGGCCTCTGTCTCGTTTTGGGAAACCAAGGTC
AGGAGGCCGTTGCAGACATAAATCTCGGCGACT
CGGCCCCGTCTCCTGAGGGTCCTGCTGGTGACCG
GCCTGGACCTTGGCCCTACAGCCCTGGAGGCCG
CTGCTGACCAGCACTGACCCCGACCTCAGAGAG
TACTCGCAGGGGCGCTGGCTGCACTCAAGACCC
TCGAGATTAACGGTGCTAACCCCGTCTGCTCCTC
CCTCCCGCAGAGACTGGGGCCTGGACTGGACAT
GAGAGCCCCTTGGTGCCACAGAGGGCTGTGTCT
TACTAGAAACAACGCAAACCTCTCCTTCCTCAGA
ATAGTGATGTGTTCGACGTTTTATCAAAGGCCCC
CTTTCTATGTTCATGTTAGTTTTGCTCCTTCTGTG
TTTTTTTCTGAACCATATCCATGTTGCTGACTTTT
CCAAATAAAGGTTTTCACTCCTCTC
3UTR-013Calr;AGAGGCCTGCCTCCAGGGCTGGACTGAGGCCTG17
calreticulinAGCGCTCCTGCCGCAGAGCTGGCCGCGCCAAAT
AATGTCTCTGTGAGACTCGAGAACTTTCATTTTT
TTCCAGGCTGGTTCGGATTTGGGGTGGATTTTGG
TTTTGTTCCCCTCCTCCACTCTCCCCCACCCCCTC
CCCGCCCTTTTTTTTTTTTTTTTTTAAACTGGTAT
TTTATCTTTGATTCTCCTTCAGCCCTCACCCCTGG
TTCTCATCTTTCTTGATCAACATCTTTTCTTGCCT
CTGTCCCCTTCTCTCATCTCTTAGCTCCCCTCCAA
CCTGGGGGGCAGTGGTGTGGAGAAGCCACAGGC
CTGAGATTTCATCTGCTCTCCTTCCTGGAGCCCA
GAGGAGGGCAGCAGAAGGGGGTGGTGTCTCCAA
CCCCCCAGCACTGAGGAAGAACGGGGCTCTTCT
CATTTCACCCCTCCCTTTCTCCCCTGCCCCCAGG
ACTGGGCCACTTCTGGGTGGGGCAGTGGGTCCC
AGATTGGCTCACACTGAGAATGTAAGAACTACA
AACAAAATTTCTATTAAATTAAATTTTGTGTCTCC
3UTR-014Colla1;CTCCCTCCATCCCAACCTGGCTCCCTCCCACCCA18
collagen,ACCAACTTTCCCCCCAACCCGGAAACAGACAAG
type I, alpha 1CAACCCAAACTGAACCCCCTCAAAAGCCAAAAA
ATGGGAGACAATTTCACATGGACTTTGGAAAAT
ATTTTTTTCCTTTGCATTCATCTCTCAAACTTAGT
TTTTATCTTTGACCAACCGAACATGACCAAAAAC
CAAAAGTGCATTCAACCTTACCAAAAAAAAAAA
AAAAAAAAGAATAAATAAATAACTTTTTAAAAA
AGGAAGCTTGGTCCACTTGCTTGAAGACCCATG
CGGGGGTAAGTCCCTTTCTGCCCGTTGGGCTTAT
GAAACCCCAATGCTGCCCTTTCTGCTCCTTTCTC
CACACCCCCCTTGGGGCCTCCCCTCCACTCCTTC
CCAAATCTGTCTCCCCAGAAGACACAGGAAACA
ATGTATTGTCTGCCCAGCAATCAAAGGCAATGCT
CAAACACCCAAGTGGCCCCCACCCTCAGCCCGC
TCCTGCCCGCCCAGCACCCCCAGGCCCTGGGGG
ACCTGGGGTTCTCAGACTGCCAAAGAAGCCTTG
CCATCTGGCGCTCCCATGGCTCTTGCAACATCTC
CCCTTCGTTTTTGAGGGGGTCATGCCGGGGGAGC
CACCAGCCCCTCACTGGGTTCGGAGGAGAGTCA
GGAAGGGCCACGACAAAGCAGAAACATCGGATT
TGGGGAACGCGTGTCAATCCCTTGTGCCGCAGG
GCTGGGCGGGAGAGACTGTTCTGTTCCTTGTGTA
ACTGTGTTGCTGAAAGACTACCTCGTTCTTGTCT
TGATGTGTCACCGGGGCAACTGCCTGGGGGCGG
GGATGGGGGCAGGGTGGAAGCGGCTCCCCATTT
TATACCAAAGGTGCTACATCTATGTGATGGGTG
GGGTGGGGAGGGAATCACTGGTGCTATAGAAAT
TGAGATGCCCCCCCAGGCCAGCAAATGTTCCTTT
TTGTTCAAAGTCTATTTTTATTCCTTGATATTTTT
CTTTTTTTTTTTTTTTTTTTGTGGATGGGGACTTG
TGAATTTTTCTAAAGGTGCTATTTAACATGGGAG
GAGAGCGTGTGCGGCTCCAGCCCAGCCCGCTGC
TCACTTTCCACCCTCTCTCCACCTGCCTCTGGCTT
CTCAGGCCTCTGCTCTCCGACCTCTCTCCTCTGA
AACCCTCCTCCACAGCTGCAGCCCATCCTCCCGG
CTCCCTCCTAGTCTGTCCTGCGTCCTCTGTCCCCG
GGTTTCAGAGACAACTTCCCAAAGCACAAAGCA
GTTTTTCCCCCTAGGGGTGGGAGGAAGCAAAAG
ACTCTGTACCTATTTTGTATGTGTATAATAATTT
GAGATGTTTTTAATTATTTTGATTGCTGGAATAA
AGCATGTGGAAATGACCCAAACATAATCCGCAG
TGGCCTCCTAATTTCCTTCTTTGGAGTTGGGGGA
GGGGTAGACATGGGGAAGGGGCTTTGGGGTGAT
GGGCTTGCCTTCCATTCCTGCCCTTTCCCTCCCCA
CTATTCTCTTCTAGATCCCTCCATAACCCCACTC
CCCTTTCTCTCACCCTTCTTATACCGCAAACCTTT
CTACTTCCTCTTTCATTTTCTATTCTTGCAATTTC
CTTGCACCTTTTCCAAATCCTCTTCTCCCCTGCAA
TACCATACAGGCAATCCACGTGCACAACACACA
CACACACTCTTCACATCTGGGGTTGTCCAAACCT
CATACCCACTCCCCTTCAAGCCCATCCACTCTCC
ACCCCCTGGATGCCCTGCACTTGGTGGCGGTGG
GATGCTCATGGATACTGGGAGGGTGAGGGGAGT
GGAACCCGTGAGGAGGACCTGGGGGCCTCTCCT
TGAACTGACATGAAGGGTCATCTGGCCTCTGCTC
CCTTCTCACCCACGCTGACCTCCTGCCGAAGGAG
CAACGCAACAGGAGAGGGGTCTGCTGAGCCTGG
CGAGGGTCTGGGAGGGACCAGGAGGAAGGCGT
GCTCCCTGCTCGCTGTCCTGGCCCTGGGGGAGTG
AGGGAGACAGACACCTGGGAGAGCTGTGGGGA
AGGCACTCGCACCGTGCTCTTGGGAAGGAAGGA
GACCTGGCCCTGCTCACCACGGACTGGGTGCCTC
GACCTCCTGAATCCCCAGAACACAACCCCCCTG
GGCTGGGGTGGTCTGGGGAACCATCGTGCCCCC
GCCTCCCGCCTACTCCTTTTTAAGCTT
3UTR-015Plod1;TTGGCCAGGCCTGACCCTCTTGGACCTTTCTTCT19
procollagen-TTGCCGACAACCACTGCCCAGCAGCCTCTGGGA
lysine, 2-CCTCGGGGTCCCAGGGAACCCAGTCCAGCCTCC
oxoglutarateTGGCTGTTGACTTCCCATTGCTCTTGGAGCCACC
5-AATCAAAGAGATTCAAAGAGATTCCTGCAGGCC
dioxygenase 1AGAGGCGGAACACACCTTTATGGCTGGGGCTCT
CCGTGGTGTTCTGGACCCAGCCCCTGGAGACAC
CATTCACTTTTACTGCTTTGTAGTGACTCGTGCTC
TCCAACCTGTCTTCCTGAAAAACCAAGGCCCCCT
TCCCCCACCTCTTCCATGGGGTGAGACTTGAGCA
GAACAGGGGCTTCCCCAAGTTGCCCAGAAAGAC
TGTCTGGGTGAGAAGCCATGGCCAGAGCTTCTC
CCAGGCACAGGTGTTGCACCAGGGACTTCTGCTT
CAAGTTTTGGGGTAAAGACACCTGGATCAGACT
CCAAGGGCTGCCCTGAGTCTGGGACTTCTGCCTC
CATGGCTGGTCATGAGAGCAAACCGTAGTCCCC
TGGAGACAGCGACTCCAGAGAACCTCTTGGGAG
ACAGAAGAGGCATCTGTGCACAGCTCGATCTTC
TACTTGCCTGTGGGGAGGGGAGTGACAGGTCCA
CACACCACACTGGGTCACCCTGTCCTGGATGCCT
CTGAAGAGAGGGACAGACCGTCAGAAACTGGA
GAGTTTCTATTAAAGGTCATTTAAACCA
3UTR-016Nucb1;TCCTCCGGGACCCCAGCCCTCAGGATTCCTGATG20
nucleobindin 1CTCCAAGGCGACTGATGGGCGCTGGATGAAGTG
GCACAGTCAGCTTCCCTGGGGGCTGGTGTCATGT
TGGGCTCCTGGGGCGGGGGCACGGCCTGGCATT
TCACGCATTGCTGCCACCCCAGGTCCACCTGTCT
CCACTTTCACAGCCTCCAAGTCTGTGGCTCTTCC
CTTCTGTCCTCCGAGGGGCTTGCCTTCTCTCGTG
TCCAGTGAGGTGCTCAGTGATCGGCTTAACTTAG
AGAAGCCCGCCCCCTCCCCTTCTCCGTCTGTCCC
AAGAGGGTCTGCTCTGAGCCTGCGTTCCTAGGTG
GCTCGGCCTCAGCTGCCTGGGTTGTGGCCGCCCT
AGCATCCTGTATGCCCACAGCTACTGGAATCCCC
GCTGCTGCTCCGGGCCAAGCTTCTGGTTGATTAA
TGAGGGCATGGGGTGGTCCCTCAAGACCTTCCC
CTACCTTTTGTGGAACCAGTGATGCCTCAAAGAC
AGTGTCCCCTCCACAGCTGGGTGCCAGGGGCAG
GGGATCCTCAGTATAGCCGGTGAACCCTGATAC
CAGGAGCCTGGGCCTCCCTGAACCCCTGGCTTCC
AGCCATCTCATCGCCAGCCTCCTCCTGGACCTCT
TGGCCCCCAGCCCCTTCCCCACACAGCCCCAGA
AGGGTCCCAGAGCTGACCCCACTCCAGGACCTA
GGCCCAGCCCCTCAGCCTCATCTGGAGCCCCTGA
AGACCAGTCCCACCCACCTTTCTGGCCTCATCTG
ACACTGCTCCGCATCCTGCTGTGTGTCCTGTTCC
ATGTTCCGGTTCCATCCAAATACACTTTCTGGAA
CAAA
3UTR-017α-globinGCTGGAGCCTCGGTGGCCATGCTTCTTGCCCCTT21
GGGCCTCCCCCCAGCCCCTCCTCCCCTTCCTGCA
CCCGTACCCCCGTGGTCTTTGAATAAAGTCTGAG
TGGGCGGC

It should be understood that those listed in the previous tables are examples and that any UTR from any gene may be incorporated into the respective first or second flanking region of the primary construct. Furthermore, multiple wild-type UTRs of any known gene may be utilized. It is also within the scope of the present invention to provide artificial UTRs which are not variants of wild type genes. These UTRs or portions thereof may be placed in the same orientation as in the transcript from which they were selected or may be altered in orientation or location. Hence a 5′ or 3′ UTR may be inverted, shortened, lengthened, made chimeric with one or more other 5′ UTRs or 3′ UTRs. As used herein, the term “altered” as it relates to a UTR sequence, means that the UTR has been changed in some way in relation to a reference sequence. For example, a 3′ or 5′ UTR may be altered relative to a wild type or native UTR by the change in orientation or location as taught above or may be altered by the inclusion of additional nucleotides, deletion of nucleotides, swapping or transposition of nucleotides. Any of these changes producing an “altered” UTR (whether 3′ or 5′) comprise a variant UTR.

In one embodiment, a double, triple or quadruple UTR such as a 5′ or 3′ UTR may be used. As used herein, a “double” UTR is one in which two copies of the same UTR are encoded either in series or substantially in series. For example, a double beta-globin 3′ UTR may be used as described in US Patent publication 20100129877, the contents of which are incorporated herein by reference in its entirety.

It is also within the scope of the present invention to have patterned UTRs. As used herein “patterned UTRs” are those UTRs which reflect a repeating or alternating pattern, such as ABABAB or AABBAABBAABB or ABCABCABC or variants thereof repeated once, twice, or more than 3 times. In these patterns, each letter, A, B, or C represent a different UTR at the nucleotide level.

In one embodiment, flanking regions are selected from a family of transcripts whose proteins share a common function, structure, feature of property. For example, oncology-related polypeptides of interest may belong to a family of proteins which are expressed in a particular cell, tissue or at some time during development. The UTRs from any of these genes may be swapped for any other UTR of the same or different family of proteins to create a new chimeric primary transcript. As used herein, a “family of proteins” is used in the broadest sense to refer to a group of two or more oncology-related polypeptides of interest which share at least one function, structure, feature, localization, origin, or expression pattern.

After optimization (if desired), the signal-sensor primary construct components are reconstituted and transformed into a vector such as, but not limited to, plasmids, viruses, cosmids, and artificial chromosomes. For example, the optimized construct may be reconstituted and transformed into chemically competent E. coli, yeast, Neurospora, maize, Drosophila, etc. where high copy plasmid-like or chromosome structures occur by methods described herein.

Stop Codons

In one embodiment, the signal-sensor primary constructs of the present invention may include at least two stop codons before the 3′ untranslated region (UTR). The stop codon may be selected from TGA, TAA and TAG. In one embodiment, the signal-sensor primary constructs of the present invention include the stop codon TGA and one additional stop codon. In a further embodiment the addition stop codon may be TAA.

Vector Amplification

The vector containing the signal-sensor primary construct is then amplified and the plasmid isolated and purified using methods known in the art such as, but not limited to, a maxi prep using the Invitrogen PURELINK™ HiPure Maxiprep Kit (Carlsbad, Calif.).

Plasmid Linearization

The plasmid may then be linearized using methods known in the art such as, but not limited to, the use of restriction enzymes and buffers. The linearization reaction may be purified using methods including, for example Invitrogen's PURELINK™ PCR Micro Kit (Carlsbad, Calif.), and HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC) and Invitrogen's standard PURELINK™ PCR Kit (Carlsbad, Calif.). The purification method may be modified depending on the size of the linearization reaction which was conducted. The linearized plasmid is then used to generate cDNA for in vitro transcription (IVT) reactions.

cDNA Template Synthesis

A cDNA template may be synthesized by having a linearized plasmid undergo polymerase chain reaction (PCR). Table 4 is a listing of primers and probes that may be useful in the PCR reactions of the present invention. It should be understood that the listing is not exhaustive and that primer-probe design for any amplification is within the skill of those in the art. Probes may also contain chemically modified bases to increase base-pairing fidelity to the target molecule and base-pairing strength. Such modifications may include 5-methyl-Cytidine, 2,6-di-amino-purine, 2′-fluoro, phosphoro-thiolate, or locked nucleic acids.

TABLE 4
Primers and Probes
Primer/SEQ
ProbeHybridizationID
IdentifierSequence (5′-3′)targetNO.
UFPTTGGACCCTCGTACAGAAGCTAAcDNA Template22
TACG
URPTx160CTTCCTACTCAGGCTTTATTCcDNA Template23
AAAGACCA
GBA1CCTTGACCTTCTGGAACTTCAcid24
glucocerebrosidase
GBA2CCAAGCACTGAAACGGATATAcid25
glucocerebrosidase
LUC1GATGAAAAGTGCTCCAAGGALuciferase26
LUC2AACCGTGATGAAAAGGTACCLuciferase27
LUC3TCATGCAGATTGGAAAGGTCLuciferase28
GCSF1CTTCTTGGACTGTCCAGAGGG-CSF29
GCSF2GCAGTCCCTGATACAAGAACG-CSF30
GCSF3GATTGAAGGTGGCTCGCTACG-CSF31
*UFP is universal forward primer; URP is universal reverse primer.

In one embodiment, the cDNA may be submitted for sequencing analysis before undergoing transcription.

Signal-Sensor Polynucleotide Production (Signal-Sensor mRNA)

The process of signal-sensor polynucleotide production may include, but is not limited to, in vitro transcription, cDNA template removal and RNA clean-up, and capping and/or tailing reactions.

In Vitro Transcription

The cDNA produced in the previous step may be transcribed using an in vitro transcription (IVT) system. The system typically comprises a transcription buffer, nucleotide triphosphates (NTPs), an RNase inhibitor and a polymerase. The NTPs may be manufactured in house, may be selected from a supplier, or may be synthesized as described herein. The NTPs may be selected from, but are not limited to, those described herein including natural and unnatural (modified) NTPs. The polymerase may be selected from, but is not limited to, T7 RNA polymerase, T3 RNA polymerase and mutant polymerases such as, but not limited to, polymerases able to be incorporated into modified nucleic acids.

RNA Polymerases

Any number of RNA polymerases or variants may be used in the design of the signal-sensor primary constructs of the present invention.

RNA polymerases may be modified by inserting or deleting amino acids of the RNA polymerase sequence. As a non-limiting example, the RNA polymerase may be modified to exhibit an increased ability to incorporate a 2′-modified nucleotide triphosphate compared to an unmodified RNA polymerase (see International Publication WO2008078180 and U.S. Pat. No. 8,101,385; herein incorporated by reference in their entireties).

Variants may be obtained by evolving an RNA polymerase, optimizing the RNA polymerase amino acid and/or nucleic acid sequence and/or by using other methods known in the art. As a non-limiting example, T7 RNA polymerase variants may be evolved using the continuous directed evolution system set out by Esvelt et al. (Nature (2011) 472(7344):499-503; herein incorporated by reference in its entirety) where clones of T7 RNA polymerase may encode at least one mutation such as, but not limited to, lysine at position 93 substituted for threonine (K93T), I4M, A7T, E63V, V64D, A65E, D66Y, T76N, C125R, S128R, A136T, N165S, G175R, H176L, Y178H, F182L, L196F, G198V, D208Y, E222K, S228A, Q239R, T243N, G259D, M267I, G280C, H300R, D351A, A354S, E356D, L360P, A383V, Y385C, D388Y, S397R, M401T, N410S, K450R, P451T, G452V, E484A, H523L, H524N, G542V, E565K, K577E, K577M, N601S, S684Y, L699I, K713E, N748D, Q754R, E775K, A827V, D851N or L864F. As another non-limiting example, T7 RNA polymerase variants may encode at least mutation as described in U.S. Pub. Nos. 20100120024 and 20070117112; herein incorporated by reference in their entireties. Variants of RNA polymerase may also include, but are not limited to, substitutional variants, conservative amino acid substitution, insertional variants, deletional variants and/or covalent derivatives.

In one embodiment, the signal-sensor primary construct may be designed to be recognized by the wild type or variant RNA polymerases. In doing so, the signal-sensor primary construct may be modified to contain sites or regions of sequence changes from the wild type or parent primary construct.

In one embodiment, the signal-sensor primary construct may be designed to include at least one substitution and/or insertion upstream of an RNA polymerase binding or recognition site, downstream of the RNA polymerase binding or recognition site, upstream of the TATA box sequence, downstream of the TATA box sequence of the signal-sensor primary construct but upstream of the coding region of the primary construct, within the 5′UTR, before the 5′UTR and/or after the 5′UTR.

In one embodiment, the 5′UTR of the signal-sensor primary construct may be replaced by the insertion of at least one region and/or string of nucleotides of the same base. The region and/or string of nucleotides may include, but is not limited to, at least 3, at least 4, at least 5, at least 6, at least 7 or at least 8 nucleotides and the nucleotides may be natural and/or unnatural. As a non-limiting example, the group of nucleotides may include 5-8 adenine, cytosine, thymine, a string of any of the other nucleotides disclosed herein and/or combinations thereof.

In one embodiment, the 5′UTR of the signal-sensor primary construct may be replaced by the insertion of at least two regions and/or strings of nucleotides of two different bases such as, but not limited to, adenine, cytosine, thymine, any of the other nucleotides disclosed herein and/or combinations thereof. For example, the 5′UTR may be replaced by inserting 5-8 adenine bases followed by the insertion of 5-8 cytosine bases. In another example, the 5′UTR may be replaced by inserting 5-8 cytosine bases followed by the insertion of 5-8 adenine bases.

In one embodiment, the signal-sensor primary construct may include at least one substitution and/or insertion downstream of the transcription start site which may be recognized by an RNA polymerase. As a non-limiting example, at least one substitution and/or insertion may occur downstream the transcription start site by substituting at least one nucleic acid in the region just downstream of the transcription start site (such as, but not limited to, +1 to +6). Changes to region of nucleotides just downstream of the transcription start site may affect initiation rates, increase apparent nucleotide triphosphate (NTP) reaction constant values, and increase the dissociation of short transcripts from the transcription complex curing initial transcription (Brieba et al, Biochemistry (2002) 41: 5144-5149; herein incorporated by reference in its entirety). The modification, substitution and/or insertion of at least one nucleic acid may cause a silent mutation of the nucleic acid sequence or may cause a mutation in the amino acid sequence.

In one embodiment, the signal-sensor primary construct may include the substitution of at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12 or at least 13 guanine bases downstream of the transcription start site.

In one embodiment, the signal-sensor primary construct may include the substitution of at least 1, at least 2, at least 3, at least 4, at least 5 or at least 6 guanine bases in the region just downstream of the transcription start site. As a non-limiting example, if the nucleotides in the region are GGGAGA the guanine bases may be substituted by at least 1, at least 2, at least 3 or at least 4 adenine nucleotides. In another non-limiting example, if the nucleotides in the region are GGGAGA the guanine bases may be substituted by at least 1, at least 2, at least 3 or at least 4 cytosine bases. In another non-limiting example, if the nucleotides in the region are GGGAGA the guanine bases may be substituted by at least 1, at least 2, at least 3 or at least 4 thymine, and/or any of the nucleotides described herein.

In one embodiment, the signal-sensor primary construct may include at least one substitution and/or insertion upstream of the start codon. For the purpose of clarity, one of skill in the art would appreciate that the start codon is the first codon of the protein coding region whereas the transcription start site is the site where transcription begins. The signal-sensor primary construct may include, but is not limited to, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 or at least 8 substitutions and/or insertions of nucleotide bases. The nucleotide bases may be inserted or substituted at 1, at least 1, at least 2, at least 3, at least 4 or at least 5 locations upstream of the start codon. The nucleotides inserted and/or substituted may be the same base (e.g., all A or all C or all T or all G), two different bases (e.g., A and C, A and T, or C and T), three different bases (e.g., A, C and T or A, C and T) or at least four different bases. As a non-limiting example, the guanine base upstream of the coding region in the signal-sensor primary construct may be substituted with adenine, cytosine, thymine, or any of the nucleotides described herein. In another non-limiting example the substitution of guanine bases in the signal-sensor primary construct may be designed so as to leave one guanine base in the region downstream of the transcription start site and before the start codon (see Esvelt et al. Nature (2011) 472(7344):499-503; herein incorporated by reference in its entirety). As a non-limiting example, at least 5 nucleotides may be inserted at 1 location downstream of the transcription start site but upstream of the start codon and the at least 5 nucleotides may be the same base type.

cDNA Template Removal and Clean-Up

The cDNA template may be removed using methods known in the art such as, but not limited to, treatment with Deoxyribonuclease I (DNase I). RNA clean-up may also include a purification method such as, but not limited to, AGENCOURT® CLEANSEQ® system from Beckman Coulter (Danvers, Mass.), HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC).

Capping and/or Tailing Reactions

The signal-sensor primary construct or mmRNA may also undergo capping and/or tailing reactions. A capping reaction may be performed by methods known in the art to add a 5′ cap to the 5′ end of the signal-sensor primary construct. Methods for capping include, but are not limited to, using a Vaccinia Capping enzyme (New England Biolabs, Ipswich, Mass.).

A poly-A tailing reaction may be performed by methods known in the art, such as, but not limited to, 2′ O-methyltransferase and by methods as described herein. If the signal-sensor primary construct generated from cDNA does not include a poly-T, it may be beneficial to perform the poly-A-tailing reaction before the signal-sensor primary construct is cleaned.

Purification

Signal-sensor primary construct or mmRNA purification may include, but is not limited to, mRNA or mmRNA clean-up, quality assurance and quality control. mRNA or mmRNA clean-up may be performed by methods known in the arts such as, but not limited to, AGENCOURT® beads (Beckman Coulter Genomics, Danvers, Mass.), poly-T beads, LNA™ oligo-T capture probes (EXIQON® Inc, Vedbaek, Denmark) or HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC). The term “purified” when used in relation to a polynucleotide such as a “purified mRNA or signal-sensor mmRNA” refers to one that is separated from at least one contaminant. As used herein, a “contaminant” is any substance which makes another unfit, impure or inferior. Thus, a purified signal-sensor polynucleotide (e.g., DNA and RNA) is present in a form or setting different from that in which it is found in nature, or a form or setting different from that which existed prior to subjecting it to a treatment or purification method.

A quality assurance and/or quality control check may be conducted using methods such as, but not limited to, gel electrophoresis, UV absorbance, or analytical HPLC.

In another embodiment, the signal-sensor mRNA or mmRNA may be sequenced by methods including, but not limited to reverse-transcriptase-PCR.

In one embodiment, the signal-sensor mRNA or mmRNA may be quantified using methods such as, but not limited to, ultraviolet visible spectroscopy (UV/Vis). A non-limiting example of a UV/Vis spectrometer is a NANODROP® spectrometer (ThermoFisher, Waltham, Mass.). The quantified signal-sensor mRNA or mmRNA may be analyzed in order to determine if the signal-sensor mRNA or mmRNA may be of proper size, check that no degradation of the signal-sensor mRNA or mmRNA has occurred. Degradation of the signal-sensor mRNA and/or mmRNA may be checked by methods such as, but not limited to, agarose gel electrophoresis, HPLC based purification methods such as, but not limited to, strong anion exchange HPLC, weak anion exchange HPLC, reverse phase HPLC (RP-HPLC), and hydrophobic interaction HPLC (HIC-HPLC), liquid chromatography-mass spectrometry (LCMS), capillary electrophoresis (CE) and capillary gel electrophoresis (CGE).

Signal Peptides or Proteins

The signal-sensor primary constructs or mmRNA may also encode additional features which facilitate trafficking of the polypeptides to therapeutically relevant sites. One such feature which aids in protein trafficking is the signal peptide sequence. As used herein, a “signal sequence” or “signal peptide” is a polynucleotide or polypeptide, respectively, which is from about 9 to 200 nucleotides (3-60 amino acids) in length which is incorporated at the 5′ (or N-terminus) of the coding region or polypeptide encoded, respectively. Addition of these sequences result in trafficking of the encoded oncology-related polypeptide to the endoplasmic reticulum through one or more secretory pathways. Some signal peptides are cleaved from the protein by signal peptidase after the proteins are transported.

Table 5 is a representative listing of signal proteins or peptides which may be incorporated for encoding by the signal-sensor polynucleotides, primary constructs or mmRNA of the invention.

TABLE 5
Signal Peptides
SEQSEQ
NUCLEOTIDE SEQUENCEIDENCODEDID
IDDescription(5′-3′)NO.PEPTIDENO.
SS-001α-1-ATGATGCCATCCTCAGTCTCA32MMPSSVS94
antitrypsinTGGGGTATTTTGCTCTTGGCGWGILLAGL
GGTCTGTGCTGTCTCGTGCCGCCLVPVSLA
GTGTCGCTCGCA
SS-002G-CSFATGGCCGGACCGGCGACTCAG33MAGPATQ95
TCGCCCATGAAACTCATGGCCSPMKLMA
CTGCAGTTGTTGCTTTGGCACLQLLLWH
TCAGCCCTCTGGACCGTCCAASALWTVQ
GAGGCGEA
SS-003Factor IXATGCAGAGAGTGAACATGATT34MQRVNMI96
ATGGCCGAGTCCCCATCGCTCMAESPSLI
ATCACAATCTGCCTGCTTGGTTICLLGYL
ACCTGCTTTCCGCCGAATGCALSAECTVF
CTGTCTTTCTGGATCACGAGALDHENAN
ATGCGAATAAGATCTTGAACCKILNRPKR
GACCCAAACGG
SS-004ProlactinATGAAAGGATCATTGCTGTTG35MKGSLLL97
CTCCTCGTGTCGAACCTTCTGLLVSNLLL
CTTTGCCAGTCCGTAGCCCCCCQSVAP
SS-005AlbuminATGAAATGGGTGACGTTCATC36MKWVTFI98
TCACTGTTGTTTTTGTTCTCGTSLLFLFSS
CCGCCTACTCCAGGGGAGTATAYSRG
TCCGCCGAVFRR
SS-006HMMSP38ATGTGGTGGCGGCTCTGGTGG37MWWRLW99
CTGCTCCTGTTGCTCCTCTTGCWLLLLLLL
TGTGGCCCATGGTGTGGGCALPMWA
MLS-ornithineTGCTCTTTAACCTCCGCATCCT38MLFNLRIL100
001carbamoyltransferaseGTTGAATAACGCTGCGTTCCGLNNAAFR
AAATGGGCATAACTTCATGGTNGHNFMV
ACGCAACTTCAGATGCGGCCARNFRCGQP
GCCACTCCAGLQ
MLS-CytochromeATGTCCGTCTTGACACCCCTG39MSVLTPLL101
002C OxidaseCTCTTGAGAGGGCTGACGGGGLRGLTGSA
subunit 8ATCCGCTAGACGCCTGCCGGTARRLPVPRA
CCGCGAGCGAAGATCCACTCCKIHSL
CTG
MLS-CytochromeATGAGCGTGCTCACTCCGTTG40MSVLTPLL102
003C OxidaseCTTCTTCGAGGGCTTACGGGALRGLTGSA
subunit 8ATCGGCTCGGAGGTTGCCCGTCRRLPVPRA
CCGAGAGCGAAGATCCATTCGKIHSL
TTG
SS-007Type III,TGACAAAAATAACTTTATCTC41MVTKITLS103
bacterialCCCAGAATTTTAGAATCCAAAPQNFRIQK
AACAGGAAACCACACTACTAQETTLLKE
AAAGAAAAATCAACCGAGAAKSTEKNSL
AAATTCTTTAGCAAAAAGTATAKSILAVK
TCTCGCAGTAAAAATCACTTCNHFIELRS
ATCGAATTAAGGTCAAAATTAKLSERFIS
TCGGAACGTTTTATTTCGCATHKNT
AAGAACACT
SS-008ViralATGCTGAGCTTTGTGGATACC42MLSFVDT104
CGCACCCTGCTGCTGCTGGCGRTLLLLAV
GTGACCAGCTGCCTGGCGACCTSCLATCQ
TGCCAG
SS-009viralATGGGCAGCAGCCAGGCGCC43MGSSQAP105
GCGCATGGGCAGCGTGGGCGRMGSVGG
GCCATGGCCTGATGGCGCTGCHGLMALL
TGATGGCGGGCCTGATTCTGCMAGLILPG
CGGGCATTCTGGCGILA
SS-010ViralATGGCGGGCATTTTTTATTTTC44MAGIFYFL106
TGTTTAGCTTTCTGTTTGGCATFSFLFGICD
TTGCGAT
SS-011ViralATGGAAAACCGCCTGCTGCGC45MENRLLR107
GTGTTTCTGGTGTGGGCGGCGVFLVWAA
CTGACCATGGATGGCGCGAGCLTMDGASA
GCG
SS-012ViralATGGCGCGCCAGGGCTGCTTT46MARQGCF108
GGCAGCTATCAGGTGATTAGCGSYQVISL
CTGTTTACCTTTGCGATTGGCFTFAIGVN
GTGAACCTGTGCCTGGGCLCLG
SS-013BacillusATGAGCCGCCTGCCGGTGCTG47MSRLPVLL109
CTGCTGCTGCAGCTGCTGGTGLLQLLVRP
CGCCCGGGCCTGCAGGLQ
SS-014BacillusATGAAACAGCAGAAACGCCT48MKQQKRL110
GTATGCGCGCCTGCTGACCCTYARLLTLL
GCTGTTTGCGCTGATTTTTCTGFALIFLLPH
CTGCCGCATAGCAGCGCGAGCSSASA
GCG
SS-015SecretionATGGCGACGCCGCTGCCTCCG49MATPLPPP111
signalCCCTCCCCGCGGCACCTGCGGSPRHLRLL
CTGCTGCGGCTGCTGCTCTCCRLLLSG
GCCCTCGTCCTCGGC
SS-016SecretionATGAAGGCTCCGGGTCGGCTC50MKAPGRL112
signalGTGCTCATCATCCTGTGCTCCVLIILCSVV
GTGGTCTTCTCTFS
SS-017SecretionATGCTTCAGCTTTGGAAACTT51MLQLWKL113
signalGTTCTCCTGTGCGGCGTGCTCLCGVLT
ACT
SS-018SecretionATGCTTTATCTCCAGGGTTGG52MLYLQGW114
signalAGCATGCCTGCTGTGGCASMPAVA
SS-019SecretionATGGATAACGTGCAGCCGAA53MDNVQPK115
signalAATAAAACATCGCCCCTTCTGIKHRPFCF
CTTCAGTGTGAAAGGCCACGTSVKGHVK
GAAGATGCTGCGGCTGGATATMLRLDIIN
TATCAACTCACTGGTAACAACSLVTTVFM
AGTATTCATGCTCATCGTATCLIVSVLALIP
TGTGTTGGCACTGATACCA
SS-020SecretionATGCCCTGCCTAGACCAACAG54MPCLDQQ116
signalCTCACTGTTCATGCCCTACCCTLTVHALPC
GCCCTGCCCAGCCCTCCTCTCPAQPSSLA
TGGCCTTCTGCCAAGTGGGGTFCQVGFLTA
TCTTAACAGCA
SS-021SecretionATGAAAACCTTGTTCAATCCA55MKTLFNP117
signalGCCCCTGCCATTGCTGACCTGAPAIADLD
GATCCCCAGTTCTACACCCTCPQFYTLSD
TCAGATGTGTTCTGCTGCAATVFCCNESE
GAAAGTGAGGCTGAGATTTTAAEILTGLT
ACTGGCCTCACGGTGGGCAGCVGSAADA
GCTGCAGATGCT
SS-022SecretionATGAAGCCTCTCCTTGTTGTG56MKPLLVV118
signalTTTGTCTTTCTTTTCCTTTGGGFVFLFLWD
ATCCAGTGCTGGCAPVLA
SS-023SecretionATGTCCTGTTCCCTAAAGTTT57MSCSLKFT119
signalACTTTGATTGTAATTTTTTTTTLIVIFFTCT
ACTGTTGGCTTTCATCCAGCLSSS
SS-024SecretionATGGTTCTTACTAAACCTCTTC58MVLTKPL120
signalAAAGAAATGGCAGCATGATGQRNGSMM
AGCTTTGAAAATGTGAAAGAASFENVKEK
AAGAGCAGAGAAGGAGGGCCSREGGPHA
CCATGCACACACACCCGAAGAHTPEEELC
AGAATTGTGTTTCGTGGTAACFVVTHTPQ
ACACTACCCTCAGGTTCAGACVQTTLNLF
CACACTCAACCTGTTTTTCCATFHIFKVLT
ATATTCAAGGTTCTTACTCAAQPLSLLWG
CCACTTTCCCTTCTGTGGGGT
SS-025SecretionATGGCCACCCCGCCATTCCGG59MATPPFRL121
signalCTGATAAGGAAGATGTTTTCCIRKMFSFK
TTCAAGGTGAGCAGATGGATGVSRWMGL
GGGCTTGCCTGCTTCCGGTCCACFRSLAAS
CTGGCGGCATCC
SS-026SecretionATGAGCTTTTTCCAACTCCTG60MSFFQLL122
signalATGAAAAGGAAGGAACTCATMKRKELIP
TCCCTTGGTGGTGTTCATGACLVVFMTV
TGTGGCGGCGGGTGGAGCCTCAAGGASS
ATCT
SS-027SecretionATGGTCTCAGCTCTGCGGGGA61MVSALRG123
signalGCACCCCTGATCAGGGTGCACAPLIRVHS
TCAAGCCCTGTTTCTTCTCCTTSPVSSPSV
CTGTGAGTGGACCACGGAGGCSGPAALVS
TGGTGAGCTGCCTGTCATCCCCLSSQSSA
AAAGCTCAGCTCTGAGCLS
SS-028SecretionATGATGGGGTCCCCAGTGAGT62MMGSPVS124
signalCATCTGCTGGCCGGCTTCTGTHLLAGFC
GTGTGGGTCGTCTTGGGCVWVVLG
SS-029SecretionATGGCAAGCATGGCTGCCGTG63MASMAAV125
signalCTCACCTGGGCTCTGGCTCTTLTWALAL
CTTTCAGCGTTTTCGGCCACCLSAFSATQA
CAGGCA
SS-030SecretionATGGTGCTCATGTGGACCAGT64MVLMWTS126
signalGGTGACGCCTTCAAGACGGCCGDAFKTA
TACTTCCTGCTGAAGGGTGCCYFLLKGAP
CCTCTGCAGTTCTCCGTGTGCLQFSVCGL
GGCCTGCTGCAGGTGCTGGTGLQVLVDL
GACCTGGCCATCCTGGGGCAGAILGQATA
GCCTACGCC
SS-031SecretionATGGATTTTGTCGCTGGAGCC65MDFVAGA127
signalATCGGAGGCGTCTGCGGTGTTIGGVCGV
GCTGTGGGCTACCCCCTGGACAVGYPLD
ACGGTGAAGGTCAGGATCCATVKVRIQT
GACGGAGCCAAAGTACACAGEPLYTGIW
GCATCTGGCACTGCGTCCGGGHCVRDTY
ATACGTATCACCGAGAGCGCGHRERVWG
TGTGGGFYRGLSLP
GCTTCTACCGGGGCCTCTCGCVCTVSLVSS
TGCCCGTGTGCACGGTGTCCC
TGGTATCTTCC
SS-032SecretionATGGAGAAGCCCCTCTTCCCA66MEKPLFPL128
signalTTAGTGCCTTTGCATTGGTTTGVPLHWFG
GCTTTGGCTACACAGCACTGGFGYTALV
TTGTTTCTGGTGGGATCGTTGVSGGIVGY
GCTATGTAAAAACAGGCAGCVKTGSVPS
GTGCCGTCCCTGGCTGCAGGGLAAGLLFG
CTGCTCTTCGGCAGTCTAGCCSLA
SS-033SecretionATGGGTCTGCTCCTTCCCCTG67MGLLLPL129
signalGCACTCTGCATCCTAGTCCTGALCILVLC
TGC
SS-034SecretionATGGGGATCCAGACGAGCCCC68MGIQTSPV130
signalGTCCTGCTGGCCTCCCTGGGGLLASLGVG
GTGGGGCTGGTCACTCTGCTCLVTLLGLA
GGCCTGGCTGTGGGCVG
SS-035SecretionATGTCGGACCTGCTACTACTG69MSDLLLL131
signalGGCCTGATTGGGGGCCTGACTGLIGGLTL
CTCTTACTGCTGCTGACGCTGLLLLTLLA
CTAGCCTTTGCCFA
SS-036SecretionATGGAGACTGTGGTGATTGTT70METVVIV132
signalGCCATAGGTGTGCTGGCCACCAIGVLATI
ATGTTTCTGGCTTCGTTTGCAGFLASFAAL
CCTTGGTGCTGGTTTGCAGGCVLVCRQ
AG
SS-037SecretionATGCGCGGCTCTGTGGAGTGC71MAGSVEC133
signalACCTGGGGTTGGGGGCACTGTTWGWGH
GCCCCCAGCCCCCTGCTCCTTCAPSPLLL
TGGACTCTACTTCTGTTTGCAWTLLLFA
GCCCCATTTGGCCTGCTGGGGAPFGLLG
SS-038SecretionATGATGCCGTCCCGTACCAAC72MMPSRTN134
signalCTGGCTACTGGAATCCCCAGTLATGIPSS
AGTAAAGTGAAATATTCAAGGKVKYSRLS
CTCTCCAGCACAGACGATGGCSTDDGYID
TACATTGACCTTCAGTTTAAGLQFKKTPP
AAAACCCCTCCTAAGATCCCTKIPYKAIA
TATAAGGCCATCGCACTTGCCLATVLFLI
ACTGTGCTGTTTTTGATTGGCGA
GCC
SS-039SecretionATGGCCCTGCCCCAGATGTGT73MALPQMC135
signalGACGGGAGCCACTTGGCCTCCDGSHLAST
ACCCTCCGCTATTGCATGACALRYCMTV
GTCAGCGGCACAGTGGTTCTGSGTVVLV
GTGGCCGGGACGCTCTGCTTCAGTLCFA
GCT
SS-041Vrg-6TGAAAAAGTGGTTCGTTGCTG74MKKWFVA136
CCGGCATCGGCGCTGCCGGACAGIGAGLL
TCATGCTCTCCAGCGCCGCCAMLSSAA
SS-042PhoAATGAAACAGAGCACCATTGCG75MKQSTIAL137
CTGGCGCTGCTGCCGCTGCTGALLPLLFT
TTTACCCCGGTGACCAAAGCGPVTKA
SS-043OmpAATGAAAAAAACCGCGATTGC76MKKTAIAI138
GATTGCGGTGGCGCTGGCGGGAVALAGF
CTTTGCGACCGTGGCGCAGGCGATVAQA
SS-044STIATGAAAAAACTGATGCTGGCG77MKKLMLA139
ATTTTTTTTAGCGTGCTGAGCTIFFSVLSFP
TTCCGAGCTTTAGCCAGAGCSFSQS
SS-045STIIATGAAAAAAAACATTGCGTTT78MKKNIAFL140
CTGCTGGCGAGCATGTTTGTGLASMFVFS
TTTAGCATTGCGACCAACGCGIATNAYA
TATGCG
SS-046AmylaseATGTTTGCGAAACGCTTTAAA79MFAKRFK141
ACCAGCCTGCTGCCGCTGTTTTSLLPLFA
GCGGGCTTTCTGCTGCTGTTTCGFLLLFHL
ATCTGGTGCTGGCGGGCCCGGVLAGPAA
CGGCGGCGAGCAS
SS-047AlphaATGCGCTTTCCGAGCATTTTT80MRFPSIFT142
FactorACCGCGGTGCTGTTTGCGGCGAVLFAASS
AGCAGCGCGCTGGCGALA
SS-048AlphaATGCGCTTTCCGAGCATTTTT81MRFPSIFT143
FactorACCACCGTGCTGTTTGCGGCGTVLFAASS
AGCAGCGCGCTGGCGALA
SS-049AlphaATGCGCTTTCCGAGCATTTTT82MRFPSIFTS144
FactorACCAGCGTGCTGTTTGCGGCGVLFAASSA
AGCAGCGCGCTGGCGLA
SS-050AlphaATGCGCTTTCCGAGCATTTTT83MRFPSIFT145
FactorACCCATGTGCTGTTTGCGGCGHVLFAASS
AGCAGCGCGCTGGCGALA
SS-051AlphaATGCGCTTTCCGAGCATTTTT84MRFPSIFTI146
FactorACCATTGTGCTGTTTGCGGCGVLFAASSA
AGCAGCGCGCTGGCGLA
SS-052AlphaATGCGCTTTCCGAGCATTTTT85MRFPSIFTF147
FactorACCTTTGTGCTGTTTGCGGCGVLFAASSA
AGCAGCGCGCTGGCGLA
SS-053AlphaATGCGCTTTCCGAGCATTTTT86MRFPSIFT148
FactorACCGAAGTGCTGTTTGCGGCGEVLFAASS
AGCAGCGCGCTGGCGALA
SS-054AlphaATGCGCTTTCCGAGCATTTTT87MRFPSIFT149
FactorACCGGCGTGCTGTTTGCGGCGGVLFAASS
AGCAGCGCGCTGGCGALA
SS-055Endoglucanase VATGCGTTCCTCCCCCCTCCTCC88MRSSPLLR150
GCTCCGCCGTTGTGGCCGCCCSAVVAAL
TGCCGGTGTTGGCCCTTGCCPVLALA
SS-056SecretionATGGGCGCGGCGGCCGTGCGC89MGAAAVR151
signalTGGCACTTGTGCGTGCTGCTGWHLCVLL
GCCCTGGGCACACGCGGGCGALGTRGRL
GCTG
SS-057FungalATGAGGAGCTCCCTTGTGCTG90MRSSLVLF152
TTCTTTGTCTCTGCGTGGACGFVSAWTA
GCCTTGGCCAGLA
SS-058FibronectinATGCTCAGGGGTCCGGGACCC91MLRGPGP153
GGGCGGCTGCTGCTGCTAGCAGRLLLLAV
GTCCTGTGCCTGGGGACATCGLCLGTSVR
GTGCGCTGCACCGAAACCGGGCTETGKSKR
AAGAGCAAGAGG
SS-059FibronectinATGCTTAGGGGTCCGGGGCCC92MLRGPGP154
GGGCTGCTGCTGCTGGCCGTCGLLLLAV
CAGCTGGGGACAGCGGTGCCCQCLGTAV
TCCACGPSTGA
SS-060FibronectinATGCGCCGGGGGGCCCTGACC93MRRGALT155
GGGCTGCTCCTGGTCCTGTGCGLLLVLCL
CTGAGTGTTGTGCTACGTGCASVVLRAAP
GCCCCCTCTGCAACAAGCAAGSATSKKRR
AAGCGCAGG

In the table, SS is secretion signal and MLS is mitochondrial leader signal. The signal-sensor primary constructs or mmRNA of the present invention may be designed to encode any of the signal peptide sequences of SEQ ID NOs 94-155, or fragments or variants thereof. These sequences may be included at the beginning of the oncology-related polypeptide coding region, in the middle or at the terminus or alternatively into a flanking region. Further, any of the signal-sensor polynucleotide primary constructs of the present invention may also comprise one or more of the sequences defined by SEQ ID NOs 32-93. These may be in the first region or either flanking region.

Additional signal peptide sequences which may be utilized in the present invention include those taught in, for example, databases such as those found at http://www.signalpeptide.de/ or http://proline.bic.nus.edu.sg/spdb/. Those described in U.S. Pat. Nos. 8,124,379; 7,413,875 and 7,385,034 are also within the scope of the invention and the contents of each are incorporated herein by reference in their entirety.

In one embodiment, the signal-sensor polynucleotide, primary constructs or mmRNA may include a nucleic acid sequence encoding a nuclear localization signal (NLS) and/or a nuclear export signal (NES). In one aspect, a signal-sensor polynucleotide, primary constructs or mmRNA may include a nucleic acid sequence encoding a nuclear localization signal (NLS). The signal-sensor polynucleotide, primary construct or mmRNA encoding a NLS would be able to traffic an oncology related polypeptide into the nucleus and deliver a survival or death signal to the nuclear microenvironment. In another aspect, the signal-sensor polynucleotide, primary constructs or mmRNA may include a nucleic acid sequence encoding a nuclear export signal such as NES1 and/or NES2. As a nonlimiting example, the signal-sensor polynucleotide, primary constructs or mmRNA may encode a NES1, NES2 and a NLS signal and an oncology related polypeptide or a scrambled sequence which is not translatable in order to interact with HIF1-alpha to alter the transcriptome of the cancer cells.

Target Selection

According to the present invention, the signal-sensor primary constructs comprise at least a first region of linked nucleosides encoding at least one oncology-related polypeptide of interest. The oncology-related polypeptides of interest or “targets” or oncology-related proteins and oncology-related peptides of the present invention are listed in Table 6, Table 7 and Table 41. Oncology-related polypeptides may be divided into classes based on their function and area of cancer intervention. For example, the classes may include targets associated with (1) apoptosis or Survival signal imbalance (AS targets). These may be caspase dependent or caspase independent targets; (2) replicative potential or anti-senescence (CC/S targets); (3) metabolic stress including the involvement of acidosis or hypoxia (O2>1%) (M targets); (4) immune response (I targets); and (5) DNA damage/protection (DDR targets).

Shown in Table 6, in addition to the name and description of the gene encoding the oncology-related polypeptide of interest are the ENSEMBL Transcript ID (ENST), the ENSEMBL Protein ID (ENSP), each present where applicable, and when available the optimized sequence ID (OPT. SEQ ID). The targets are also categorized by group where “AS” refers to targets involved in apoptotic signaling; “M” refers to targets involved in metabolic processes and “CC/S” refers to targets involved in cell cycle and senescense.

TABLE 6
Oncology Related Targets
Prot.OPT.
Trans.SEQSEQ
ENSTSEQENSPIDID
Cat.TargetTarget DescriptionIDID NOIDNONO
AS14-3-3tyrosine 3-2380811562380811321
monooxygenase/tryptophan 5-
monooxygenase activation
protein, theta polypeptide
AS14-3-3tyrosine 3-2489751572489751322
monooxygenase/tryptophan 5-
monooxygenase activation
protein, eta polypeptide
AS14-3-3tyrosine 3-2643351582643351323
monooxygenase/tryptophan 5-
monooxygenase activation
protein, epsilon polypeptide
AS14-3-3tyrosine 3-3076301593063301324
monooxygenase/tryptophan 5-
monooxygenase activation
protein, gamma polypeptide
AS14-3-3tyrosine 3-3532451603095031325
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-3537031613001611326
monooxygenase/tryptophan 5-
monooxygenase activation
protein, beta polypeptide
AS14-3-3tyrosine 3-3728391623619301327
monooxygenase/tryptophan 5-
monooxygenase activation
protein, beta polypeptide
AS14-3-3tyrosine 3-3818441633712671328
monooxygenase/tryptophan 5-
monooxygenase activation
protein, theta polypeptide
AS14-3-3tyrosine 3-3959481643792781329
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-3959511653792811330
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-3959531663792831331
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-3959561673792861332
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-3959571683792871333
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-3959581693792881334
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-4141311704060581335
monooxygenase/tryptophan 5-
monooxygenase activation
protein, epsilon polypeptide
AS14-3-3tyrosine 3-4189971714165511336
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-4194771723951141337
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-4282621733947291338
monooxygenase/tryptophan 5-
monooxygenase activation
protein, beta polypeptide
AS14-3-3tyrosine 3-4372931743948801339
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-4458301753945581340
monooxygenase/tryptophan 5-
monooxygenase activation
protein, beta polypeptide
AS14-3-3tyrosine 3-4466191763989901341
monooxygenase/tryptophan 5-
monooxygenase activation
protein, theta polypeptide
AS14-3-3tyrosine 3-4532071773906451342
monooxygenase/tryptophan 5-
monooxygenase activation
protein, gamma polypeptide
AS14-3-3tyrosine 3-4573091783985991343
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5177971794278011344
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5213091804296231345
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5213281814290411346
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5216071824300581347
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5225421834300721348
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5228191844287751349
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5231311854283811350
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5238481864288601351
monooxygenase/tryptophan 5-
monooxygenase activation
protein, zeta polypeptide
AS14-3-3tyrosine 3-5367551874438031352
monooxygenase/tryptophan 5-
monooxygenase activation
protein, gamma polypeptide
AS14-3-3tyrosine 3-5399791884432261353
monooxygenase/tryptophan 5-
monooxygenase activation
protein, theta polypeptide
ASAIFapoptosis-inducing factor,2872951892872951354
mitochondrion-associated, 1
ASAIFapoptosis-inducing factor,3078641903123701355
mitochondrion-associated, 2
ASAIFapoptosis-inducing factor,3199081913151221356
mitochondrion-associated, 1
ASAIFapoptosis-inducing factor,3336071923276711357
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,3353751933353691358
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,3464241943163201359
mitochondrion-associated, 1
ASAIFapoptosis-inducing factor,3732481953623451360
mitochondrion-associated, 2
ASAIFapoptosis-inducing factor,3950391963784801361
mitochondrion-associated, 2
ASAIFapoptosis-inducing factor,3991631973821161362
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,3991671983821201363
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,4050891993858001364
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,4347142003996571365
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,4402382013907981366
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,4402632024058791367
mitochondrion-associated, 1
ASAIFapoptosis-inducing factor,4413762034020671368
mitochondrion-associated, 3
ASAIFapoptosis-inducing factor,4604362044312221369
mitochondrion-associated, 1
ASAIFapoptosis-inducing factor,5357242054461131370
mitochondrion-associated, 1
ASAKTv-akt murine thymoma viral2638262062638261371
(PKB)oncogene hom*olog 3 (protein
kinase B, gamma)
ASAKTv-akt murine thymoma viral3112782073094281372
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral3361992083369431373
(PKB)oncogene hom*olog 3 (protein
kinase B, gamma)
ASAKTv-akt murine thymoma viral3493102092702021374
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral3583352103510951375
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral3665392113554971376
(PKB)oncogene hom*olog 3 (protein
kinase B, gamma)
ASAKTv-akt murine thymoma viral3665402123554981377
(PKB)oncogene hom*olog 3 (protein
kinase B, gamma)
ASAKTv-akt murine thymoma viral3918442133757191378
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral3920372143758911379
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral3920382153758921380
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4026152163853261381
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral4077962173842931382
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral4163622184079991383
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4169942193924581384
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4231272204038421385
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4249012213995321386
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4273752224038901387
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4520772234040831388
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral4564412243965321389
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral5378342254415911390
(PKB)oncogene hom*olog 2
ASAKTv-akt murine thymoma viral5441682264438971391
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral5526312274478201392
(PKB)oncogene hom*olog 3 (protein
kinase B, gamma)
ASAKTv-akt murine thymoma viral5545812284518281393
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral5548482294511661394
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral5555282304506881395
(PKB)oncogene hom*olog 1
ASAKTv-akt murine thymoma viral5559262314518241396
(PKB)oncogene hom*olog 1
ASANTsolute carrier family 252814562322814561397
(mitochondrial carrier; adenine
nucleotide translocator),
member 4
ASApaf-1apoptotic peptidase activating3339912333345581398
factor 1
ASApaf-1apoptotic peptidase activating3394332343418301399
factor 1
ASApaf-1apoptotic peptidase activating3573102353498621400
factor 1
ASApaf-1apoptotic peptidase activating3599722363530591401
factor 1
ASApaf-1apoptotic peptidase activating5470452374497911402
factor 1
ASApaf-1apoptotic peptidase activating5490072384481611403
factor 1
ASApaf-1apoptotic peptidase activating5505272394484491404
factor 1
ASApaf-1apoptotic peptidase activating5519642404481651405
factor 1
ASApaf-1apoptotic peptidase activating5522682414488261406
factor 1
ASAPRILtumor necrosis factor (ligand)3387842423435051407
(TNFSF13)superfamily, member 13
ASAPRILtumor necrosis factor (ligand)3492282433144551408
(TNFSF13)superfamily, member 13
ASAPRILtumor necrosis factor (ligand)3805352443699081409
(TNFSF13)superfamily, member 13
ASAPRILtumor necrosis factor (ligand)3965452453797941410
(TNFSF13)superfamily, member 13
ASARTSphosphoribosyl pyrophosphate3724182463614951411
synthetase 1
ASARTSphosphoribosyl pyrophosphate3724192473614961412
synthetase 1
ASARTSphosphoribosyl pyrophosphate3724282483615051413
synthetase 1
ASARTSphosphoribosyl pyrophosphate3724352493615121414
synthetase 1
ASARTSphosphoribosyl pyrophosphate5432482504431851415
synthetase 1
ASASK1mitogen-activated protein3558452513481041416
(MAP3K5)kinase kinase kinase 5
ASASK1mitogen-activated protein3590152523519081417
(MAP3K5)kinase kinase kinase 5
ASASK1mitogen-activated protein3677682533567421418
(MAP3K5)kinase kinase kinase 5
ASBADBCL2-associated agonist of3090322543091031419
cell death
ASBADBCL2-associated agonist of3945322553780401420
cell death
ASBADBCL2-associated agonist of5401522564408071421
cell death
ASBAFF(TNFSF13B)tumor necrosis factor (ligand)3758872573650481422
superfamily, member 13b
ASBAFF(TNFSF13B)tumor necrosis factor (ligand)4305592583895401423
superfamily, member 13b
ASBAFF(TNFSF13B)tumor necrosis factor (ligand)5421362594453341424
superfamily, member 13b
ASBakBCL2-antagonist/killer 13606612603538781425
ASBakBCL2-antagonist/killer 13744602613635841426
ASBakBCL2-antagonist/killer 13744672623635911427
ASBakBCL2-antagonist/killer 14429982633912581428
ASBAXBCL2-associated X protein2932882642932881429
ASBAXBCL2-associated X protein3453582652632621430
ASBAXBCL2-associated X protein3544702663464611431
ASBAXBCL2-associated X protein3918712673757441432
ASBAXBCL2-associated X protein4159692683899711433
ASBAXBCL2-associated X protein5397872694414131434
ASBcl-2B-cell CLL/lymphoma 23336812703296231435
ASBcl-2B-cell CLL/lymphoma 23981172713811851436
ASBcl-2B-cell CLL/lymphoma 24444842724042141437
ASBcl-BBCL2-like 10 (apoptosis2604422732604421438
facilitator)
ASBcl-WBCL2-like 22504052742504051439
ASBcl-WBCL2-like 25546352754512341440
ASBcl-WBCL2-like 25572362764517011441
ASBcl-WBCL2-like 25575792774522651442
ASBcl-XLBCL2-like 13076772783025641443
ASBcl-XLBCL2-like 13760552793652231444
ASBcl-XLBCL2-like 13760622803652301445
ASBcl-XLBCL2-like 14204882813907601446
ASBcl-XLBCL2-like 14206532824055631447
ASBcl-XLBCL2-like 14229202834112521448
ASBcl-XLBCL2-like 14392672843896881449
ASBcl-XLBCL2-like 14502732854062031450
ASBcl-XLBCL2-like 14564042863955451451
ASBCMAtumor necrosis factor receptor 53243287 532431452
superfamily, member 17
ASBCMAtumor necrosis factor receptor3964952883797531453
superfamily, member 17
ASBCMAtumor necrosis factor receptor4353552894017821454
superfamily, member 17
ASBFL1BCL2-related protein A12679532902679531455
ASBFL1BCL2-related protein A13356612913352501456
ASBidBH3 interacting domain death3173612923188221457
agonist
ASBidBH3 interacting domain death3421112933445941458
agonist
ASBidBH3 interacting domain death3997652943826671459
agonist
ASBidBH3 interacting domain death3997672953826691460
agonist
ASBidBH3 interacting domain death3997742963826741461
agonist
ASBidBH3 interacting domain death5519522974492361462
agonist
ASBikBCL2-interacting killer2161152982161151463
(apoptosis-inducing)
ASBimBCL2-like 11 (apoptosis3086592993092261464
facilitator)
ASBimBCL2-like 11 (apoptosis3375653003383741465
facilitator)
ASBimBCL2-like 11 (apoptosis3577573013503981466
facilitator)
ASBimBCL2-like 11 (apoptosis3932523023769411467
facilitator)
ASBimBCL2-like 11 (apoptosis3932533033769421468
facilitator)
ASBimBCL2-like 11 (apoptosis3932563043769431469
facilitator)
ASBimBCL2-like 11 (apoptosis4321793054118701470
facilitator)
ASBimBCL2-like 11 (apoptosis4520333064036661471
facilitator)
ASBMFBcl2 modifying factor2204463072204461472
ASBMFBcl2 modifying factor3546703083466971473
ASBMFBcl2 modifying factor3975733093807031474
ASBMFBcl2 modifying factor4314153103965111475
ASBMFBcl2 modifying factor5597013114539191476
ASBMFBcl2 modifying factor5612823124535221477
ASBMFBcl2 modifying factor5613603134538921478
ASBREbrain and reproductive organ-3420453143393711479
expressed (TNFRSF1A
modulator)
ASBREbrain and reproductive organ-3447733153434121480
expressed (TNFRSF1A
modulator)
ASBREbrain and reproductive organ-3617043163546991481
expressed (TNFRSF1A
modulator)
ASBREbrain and reproductive organ-3796233173689441482
expressed (TNFRSF1A
modulator)
ASBREbrain and reproductive organ-3796243183689451483
expressed (TNFRSF1A
modulator)
ASBREbrain and reproductive organ-3796323193689531484
expressed (TNFRSF1A
modulator)
ASBREbrain and reproductive organ-4369243203923451485
expressed (TNFRSF1A
modulator)
ASCalcineurin Aprotein phosphatase 3, catalytic3230553213205801486
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic3948533223783221487
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic3948543233783231488
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic5071763244229901489
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic5122153254227811490
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic5236943264293501491
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic5258193274345991492
subunit, alpha isozyme
ASCalcineurin Aprotein phosphatase 3, catalytic5293243284316191493
subunit, alpha isozyme
ASCaspase-1caspase 1, apoptosis-related3532473293441321494
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related3931363303768441495
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related4159813314084461496
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related4368633324100761497
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related4463693334032601498
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5258253344347791499
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5265683354342501500
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5289743364342591501
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5298713374319471502
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5311663384343031503
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5334003394331381504
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-1caspase 1, apoptosis-related5344973404368751505
cysteine peptidase (interleukin
1, beta, convertase)
ASCaspase-caspase 10, apoptosis-related2728793412728791506
10cysteine peptidase
ASCaspase-caspase 10, apoptosis-related2861863422861861507
10cysteine peptidase
ASCaspase-caspase 10, apoptosis-related3468173432378651508
10cysteine peptidase
ASCaspase-caspase 10, apoptosis-related3601323443532501509
10cysteine peptidase
ASCaspase-2caspase 2, apoptosis-related3104473453126641510
cysteine peptidase
ASCaspase-2caspase 2, apoptosis-related3506233463400301511
cysteine peptidase
ASCaspase-2caspase 2, apoptosis-related3929233473766541512
cysteine peptidase
ASCaspase-3caspase 3, apoptosis-related3083943483110321513
cysteine peptidase
ASCaspase-3caspase 3, apoptosis-related4384673493907921514
cysteine peptidase
ASCaspase-3caspase 3, apoptosis-related4471213504071421515
cysteine peptidase
ASCaspase-3caspase 3, apoptosis-related5239163514289291516
cysteine peptidase
ASCaspase-4caspase 4, apoptosis-related3555463523477411517
cysteine peptidase
ASCaspase-4caspase 4, apoptosis-related4174403534016731518
cysteine peptidase
ASCaspase-4caspase 4, apoptosis-related4447393543885661519
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related2603153552603151520
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related3931393563768471521
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related3931413573768491522
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related4184343583981301523
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related4447493593883651524
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related5260563604368771525
cysteine peptidase
ASCaspase-5caspase 5, apoptosis-related5313673614344711526
cysteine peptidase
ASCaspase-6caspase 6, apoptosis-related2651643622651641527
cysteine peptidase
ASCaspase-6caspase 6, apoptosis-related3529813632853331528
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3456333642987011529
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3693153653583211530
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3693163663583221531
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3693183673583241532
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3693193683583251533
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3693213693583271534
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related3693313703583371535
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related4296173714000941536
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related4423933723944821537
cysteine peptidase
ASCaspase-7caspase 7, apoptosis-related4524903733981071538
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related2642743742642741539
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related2642753752642751540
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3234923763257221541
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3584853773512731542
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3922583783760871543
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3922593793760881544
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3922613803760891545
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3922633813760911546
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related3922663823760941547
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related4137263833975281548
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related4298813843906411549
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related4321093854125231550
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related4407323863968691551
cysteine peptidase
ASCaspase-8caspase 8, apoptosis-related4476163873883061552
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related3338683883302371553
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related3485493892552561554
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related3758743903650341555
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related3758903913650511556
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related4404843924113041557
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related4475223933965401558
cysteine peptidase
ASCaspase-9caspase 9, apoptosis-related5464243944495841559
cysteine peptidase
ASCD27CD27 molecule2665573952665571560
ASCD30tumor necrosis factor receptor2639323962639321561
superfamily, member 8
ASCD30tumor necrosis factor receptor4131463973983371562
superfamily, member 8
ASCD30tumor necrosis factor receptor4178143983906501563
superfamily, member 8
ASCD30Ltumor necrosis factor (ligand)2237953992237951564
superfamily, member 8
ASCD40CD40 molecule, TNF receptor3722784003613521565
superfamily member 5
ASCD40LCD40 ligand3706284013596621566
(TNFSF5)
ASCD40LCD40 ligand3706294023596631567
(TNFSF5)
ASCD41CD40 molecule, TNF receptor3722764033613501568
superfamily member 5
ASCD42CD40 molecule, TNF receptor3722854043613591569
superfamily member 5
ASCD70(TNFSF7)CD70 molecule2459034052459031570
ASCD70(TNFSF7)CD70 molecule4231454063952941571
ASCDK1cyclin-dependent kinase 13166294073259701572
(p34)
ASCDK1cyclin-dependent kinase 13738094083629151573
(p34)
ASCDK1cyclin-dependent kinase 13952844093786991574
(p34)
ASCDK1cyclin-dependent kinase 14482574103979731575
(p34)
ASCDK1cyclin-dependent kinase 15190784114306651576
(p34)
ASCDK5cyclin-dependent kinase 54859724124197821577
ASCDK5R1cyclin-dependent kinase 5,3134014133184861578
(p35)regulatory subunit 1 (p35)
ASc-CASP8 and FADD-like3099554143124551579
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like3408704153393261580
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like3433754163393911581
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like3555584173477571582
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like3951484183785801583
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like4177484194128821584
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like4232414203994201585
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like4334454213910291586
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like4412244224118971587
FLIP(S)apoptosis regulator
ASc-CASP8 and FADD-like4432274234132701588
FLIP(S)apoptosis regulator
AScIAP1baculoviral IAP repeatNA424NA15892488
containing 3
ASc-IAP1baculoviral IAP repeat2634644252634641590
containing 3
ASc-IAP1baculoviral IAP repeat5328084264329071591
containing 3
AScIAP2baculoviral IAP repeatNA427NA1592
containing 2
ASC-IAP2baculoviral IAP repeat2277584282277581593
containing 2
ASC-IAP2baculoviral IAP repeat5306754294317231594
containing 2
ASC-IAP2baculoviral IAP repeat5326724304349791595
containing 2
ASC-IAP2baculoviral IAP repeat5417414314407711596
containing 2
ASc-Junjun proto-oncogene3712224323602661597
ASc-Raf-1v-raf-1 murine leukemia viral2518494332518491598
oncogene hom*olog 1
ASc-Raf-1v-raf-1 murine leukemia viral4424154344018881599
oncogene hom*olog 1
ASc-Raf-1v-raf-1 murine leukemia viral5349974354411861600
oncogene hom*olog 1
ASc-Raf-1v-raf-1 murine leukemia viral5421774364435671601
oncogene hom*olog 1
ASCytochrome ccytochrome c, somatic3057864373077861602
ASCytochrome ccytochrome c, somatic4094094383862701603
ASCytochrome ccytochrome c, somatic4097644393872791604
ASCytochrome ccytochrome c, somatic4134474404164791605
ASDAXXdeath-domain associated2660004412660001606
protein
ASDAXXdeath-domain associated3745424423636681607
protein
ASDAXXdeath-domain associated3830624433725391608
protein
ASDAXXdeath-domain associated3831944443726811609
protein
ASDAXXdeath-domain associated3990604453820141610
protein
ASDAXXdeath-domain associated3993444463822811611
protein
ASDAXXdeath-domain associated4140834473968761612
protein
ASDAXXdeath-domain associated4142724484097561613
protein
ASDAXXdeath-domain associated4198554493976121614
protein
ASDAXXdeath-domain associated4282684504082151615
protein
ASDAXXdeath-domain associated4295314514158981616
protein
ASDAXXdeath-domain associated4334824524046231617
protein
ASDAXXdeath-domain associated4363114534043761618
protein
ASDAXXdeath-domain associated4383324544117001619
protein
ASDAXXdeath-domain associated4405004554039861620
protein
ASDAXXdeath-domain associated4450094563941081621
protein
ASDAXXdeath-domain associated4464034574060081622
protein
ASDAXXdeath-domain associated4534074584084991623
protein
ASDAXXdeath-domain associated4539314594124331624
protein
ASDAXXdeath-domain associated4541974604121771625
protein
ASDAXXdeath-domain associated4558604614107721626
protein
ASDAXXdeath-domain associated5476634624471151627
protein
ASDAXXdeath-domain associated5486044634483371628
protein
ASDAXXdeath-domain associated5508224644478611629
protein
ASDAXXdeath-domain associated5529444654478331630
protein
ASDcR3tumor necrosis factor receptor3428524663423281631
superfamily, member 6b,
decoy
ASDcR3tumor necrosis factor receptor3699964673590131632
superfamily, member 6b,
decoy
ASDcR3tumor necrosis factor receptor3700064683590231633
superfamily, member 6b,
decoy
ASDFF40DNA fragmentation factor,3388954693395241634
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,3393504703432181635
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,3413854713459061636
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,3782064723674481637
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,3782094733674541638
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,3782124743674571639
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,4305394753895021640
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,4486324764116351641
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDFF40DNA fragmentation factor,4919984774367751642
(CAD)40 kDa, beta polypeptide
(caspase-activated DNase)
ASDR3tumor necrosis factor receptor3483334783144511643
superfamily, member 25
ASDR3tumor necrosis factor receptor3517484793267621644
superfamily, member 25
ASDR3tumor necrosis factor receptor3519594803377131645
superfamily, member 25
ASDR3tumor necrosis factor receptor3568764813493411646
superfamily, member 25
ASDR3tumor necrosis factor receptor3777824823670131647
superfamily, member 25
ASDR4tumor necrosis factor receptor2211324832211321648
superfamily, member 10a
ASDR5tumor necrosis factor receptor2764314842764311649
superfamily, member 10b
ASDR5tumor necrosis factor receptor3477394853178591650
superfamily, member 10b
ASDR5tumor necrosis factor receptor5422264864433861651
superfamily, member 10b
ASDR6tumor necrosis factor receptor2968614872968611652
superfamily, member 21
ASDR6tumor necrosis factor receptor4192064883900321653
superfamily, member 21
ASEGFRepidermal growth factor2754934892754931654
receptor
ASEGFRepidermal growth factor3429164903423761655
receptor
ASEGFRepidermal growth factor3445764913459731656
receptor
ASEGFRepidermal growth factor3955044923788801657
receptor
ASEGFRepidermal growth factor4203164934138431658
receptor
ASEGFRepidermal growth factor4425914944100311659
receptor
ASEGFRepidermal growth factor4547574953952431660
receptor
ASEGFRepidermal growth factor4550894964155591661
receptor
ASEGFRepidermal growth factor5334504974352621662
receptor
ASErbB2v-erb-b2 erythroblastic2695714982695711663
leukemia viral oncogene
hom*olog 2, neuro/glioblastoma
derived oncogene hom*olog
(avian)
ASErbB2v-erb-b2 erythroblastic4063814993851851664
leukemia viral oncogene
hom*olog 2, neuro/glioblastoma
derived oncogene hom*olog
(avian)
ASErbB2v-erb-b2 erythroblastic4456585004040471665
leukemia viral oncogene
hom*olog 2, neuro/glioblastoma
derived oncogene hom*olog
(avian)
ASErbB2v-erb-b2 erythroblastic5400425014463821666
leukemia viral oncogene
hom*olog 2, neuro/glioblastoma
derived oncogene hom*olog
(avian)
ASErbB2v-erb-b2 erythroblastic5401475024435621667
leukemia viral oncogene
hom*olog 2, neuro/glioblastoma
derived oncogene hom*olog
(avian)
ASErbB2v-erb-b2 erythroblastic5417745034464661668
leukemia viral oncogene
hom*olog 2, neuro/glioblastoma
derived oncogene hom*olog
(avian)
ASErbB3v-erb-b2 erythroblastic2671015042671011669
leukemia viral oncogene
hom*olog 3 (avian)
ASErbB3v-erb-b2 erythroblastic3940995053776591670
leukemia viral oncogene
hom*olog 3 (avian)
ASErbB3v-erb-b2 erythroblastic4117315064157531671
leukemia viral oncogene
hom*olog 3 (avian)
ASErbB3v-erb-b2 erythroblastic4152885074083401672
leukemia viral oncogene
hom*olog 3 (avian)
ASErbB3v-erb-b2 erythroblastic4501465083991781673
leukemia viral oncogene
hom*olog 3 (avian)
ASErbB3v-erb-b2 erythroblastic5492825094486361674
leukemia viral oncogene
hom*olog 3 (avian)
ASErbB3v-erb-b2 erythroblastic5510855104484831675
leukemia viral oncogene
hom*olog 3 (avian)
ASErk(MAPK1/mitogen-activated protein2158325112158321676
3)kinase 1
ASErk(MAPK1/mitogen-activated protein2630255122630251677
3)kinase 3
ASErk(MAPK1/mitogen-activated protein3222665133272931678
3)kinase 3
ASErk(MAPK1/mitogen-activated protein3952005143786261679
3)kinase 3
ASErk(MAPK1/mitogen-activated protein3952025153786281680
3)kinase 3
ASErk(MAPK1/mitogen-activated protein3988225163818031681
3)kinase 1
ASErk(MAPK1/mitogen-activated protein4033945173848951682
3)kinase 3
ASErk(MAPK1/mitogen-activated protein4159115184091491683
3)kinase 1
ASErk(MAPK1/mitogen-activated protein4846635194327421684
3)kinase 3
ASErk(MAPK1/mitogen-activated protein5447865204408421685
3)kinase 1
ASFADDFas (TNFRSF6)-associated via3018385213018381686
death domain
ASFLASHcaspase 8 associated protein 2237177522NA1687
ASFLASHcaspase 8 associated protein 2419040523NA
ASFLASHcaspase 8 associated protein 2444163524NA
ASFLASHcaspase 8 associated protein 2547893525NA
ASFLASHcaspase 8 associated protein 2548224526NA
ASFLASHcaspase 8 associated protein 2551025527NA
ASFLASHcaspase 8 associated protein 2552401528NA
ASFN14tumor necrosis factor receptor3265775293267371688
superfamily, member 12A
ASFN14tumor necrosis factor receptor3416275303438941689
superfamily, member 12A
ASGCKmitogen-activated protein2940665312940661690
(MAP4K2)kinase kinase kinase kinase 2
ASGRB2growth factor receptor-bound3166155323173601691
protein 2
ASGRB2growth factor receptor-bound3168045333390071692
protein 2
ASGRB2growth factor receptor-bound3925625343763451693
protein 2
ASGRB2growth factor receptor-bound3925645353763471694
protein 2
ASH-Rasv-Ha-ras Harvey rat sarcoma3111895363098451695
viral oncogene hom*olog
ASH-Rasv-Ha-ras Harvey rat sarcoma3975945373807221696
viral oncogene hom*olog
ASH-Rasv-Ha-ras Harvey rat sarcoma3975965383807231697
viral oncogene hom*olog
ASH-Rasv-Ha-ras Harvey rat sarcoma4173025393882461698
viral oncogene hom*olog
ASH-Rasv-Ha-ras Harvey rat sarcoma4515905404075861699
viral oncogene hom*olog
ASH-Rasv-Ha-ras Harvey rat sarcoma4932305414340231700
viral oncogene hom*olog
ASHRKharakiri, BCL2 interacting2575725422575721701
protein (contains only BH3
domain)
ASHSP27heat shock 27 kDa protein 12485535432485531702
ASHSP27heat shock 27 kDa protein 33020055443033941703
ASHSP27Heat shock protein beta-23042985453024761704
ASHSP27heat shock 27 kDa protein 14322765464065451705
ASHSP27Heat shock protein beta-25373825474455851706
ASHtrA2/OmiHtrA serine peptidase 22580805482580801707
ASHtrA2/OmiHtrA serine peptidase 23522225493128931708
ASHumaninMT-RNR2-like 43999745503828561709
ASHumaninMT-RNR2-like 55125245514379101710
ASHumaninMT-RNR2-like 85366845524396661711
ASHumaninMT-RNR2-like 15400405534392281712
ASHumaninMT-RNR2-like 35435005544433391713
ASHumaninMT-RNR2-like 75448245554399851714
ASHumaninMT-RNR2-like 105450755564421591715
ASHumaninMT-RNR2-like 65704195574610751716
ASICADDNA fragmentation factor,3770365583662351717
45 kDa, alpha polypeptide
ASICADDNA fragmentation factor,3770385593662371718
45 kDa, alpha polypeptide
ASIGF-1Rinsulin-like growth factor 12680355602680351719
receptor
ASIKKconserved helix-loop-helix3703975613594241720
(alpha)ubiquitous kinase
ASIKKinhibitor of kappa light3797085623690301721
(beta)polypeptide gene enhancer in
B-cells, kinase beta
ASIKKinhibitor of kappa light4165055634049201722
(beta)polypeptide gene enhancer in
B-cells, kinase beta
ASIKKinhibitor of kappa light5208105644306841723
(beta)polypeptide gene enhancer in
B-cells, kinase beta
ASIKK-inhibitor of kappa light2635185652635181724
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light3696015663586141725
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light3696065673586191726
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light3696075683586201727
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light3696095693586221728
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light4226805703903681729
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light4402865713949341730
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light4456225723952051731
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIKK-inhibitor of kappa light4555885734007691732
gammapolypeptide gene enhancer in
B-cells, kinase gamma
ASIRAK1interleukin-1 receptor-3699805743589971733
associated kinase 1
ASIRAK1interleukin-1 receptor-3936825753772871734
associated kinase 1
ASIRAK1interleukin-1 receptor-3936875763772911735
associated kinase 1
ASIRAK1interleukin-1 receptor-4299365773926621736
associated kinase 1
ASIRAK2interleukin-1 receptor-2564585782564581737
associated kinase 2
ASIRS-1insulin receptor substrate 13051235793048951738
ASjBid;jBIDNANA1739
formed
after
cleaving
BID at
position
25
ASJNK1(MAPK8)mitogen-activated protein3603325803534831740
kinase 8
ASJNK1(MAPK8)mitogen-activated protein3741745813632891741
kinase 8
ASJNK1(MAPK8)mitogen-activated protein3741765823632911742
kinase 8
ASJNK1(MAPK8)mitogen-activated protein3741795833632941743
kinase 8
ASJNK1(MAPK8)mitogen-activated protein3741825843632971744
kinase 8
ASJNK1(MAPK8)mitogen-activated protein3741895853633041745
kinase 8
ASJNK1(MAPK8)mitogen-activated protein3956115863789741746
kinase 8
ASJNK1(MAPK8)mitogen-activated protein4265575873977291747
kinase 8
ASJNK1(MAPK8)mitogen-activated protein4290415883932231748
kinase 8
ASJNK1(MAPK8)mitogen-activated protein4323795893879361749
kinase 8
ASJNK3(MAPK10)mitogen-activated protein3592215903521571750
kinase 10
ASJNK3(MAPK10)mitogen-activated protein3615695913552971751
kinase 10
ASJNK3(MAPK10)mitogen-activated protein3951575923785861752
kinase 10
ASJNK3(MAPK10)mitogen-activated protein3951605933785891753
kinase 10
ASJNK3(MAPK10)mitogen-activated protein3951615943785901754
kinase 10
ASJNK3(MAPK10)mitogen-activated protein3951665953785951755
kinase 10
ASJNK3(MAPK10)mitogen-activated protein3951695963785981756
kinase 10
ASJNK3(MAPK10)mitogen-activated protein4490475974144691757
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5023025984239181758
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5039115994214091759
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5067736004213591760
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5094646014241281761
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5111676024222771762
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5113286034217621763
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5120176044247551764
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5125646054229851765
kinase 10
ASJNK3(MAPK10)mitogen-activated protein5154006064241541766
kinase 10
ASMAP1mannan-binding lectin serine1692936071692931767
peptidase 1 (C4/C2 activating
component of Ra-reactive
factor)
ASMAP1mannan-binding lectin serine2962806082962801768
peptidase 1 (C4/C2 activating
component of Ra-reactive
factor)
ASMAP1mannan-binding lectin serine3377746093367921769
peptidase 1 (C4/C2 activating
component of Ra-reactive
factor)
ASMAP1mannan-binding lectin serine3924726103762641770
peptidase 1 (C4/C2 activating
component of Ra-reactive
factor)
ASMAP1mannan-binding lectin serine5418116114404461771
peptidase 1 (C4/C2 activating
component of Ra-reactive
factor)
ASMAP1mannan-binding lectin serine5418966124462401772
peptidase 1 (C4/C2 activating
component of Ra-reactive
factor)
ASMcl-1myeloid cell leukemia3079406133099731773
sequence 1 (BCL2-related)
ASMcl-1myeloid cell leukemia3690266143580221774
sequence 1 (BCL2-related)
ASMcl-1myeloid cell leukemia4397496154113951775
sequence 1 (BCL2-related)
ASMEK1mitogen-activated protein2158326162158321776
(MAP2K1)kinase 1
ASMEK1mitogen-activated protein3071026173024861777
(MAP2K1)kinase kinase 1
ASMEK1mitogen-activated protein4159116184091491778
(MAP2K1)kinase 1
ASMEK1mitogen-activated protein5447866194408421779
(MAP2K1)kinase 1
ASMEK2mitogen-activated protein2629486202629481780
(MAP2K2)kinase kinase 2
ASMEK4mitogen-activated protein3535336212624451781
(MAP2K4)kinase kinase 4
ASMEK4mitogen-activated protein4153856224104021782
(MAP2K4)kinase kinase 4
ASMEK4mitogen-activated protein5364136234416101783
(MAP2K4)kinase kinase 4
ASMEK4mitogen-activated protein5384656244448741784
(MAP2K4)kinase kinase 4
ASMEKK1mitogen-activated protein3995036253824231785
(MAP3K1)kinase kinase kinase 1
ASNADEnerve growth factor receptor2998726262998721786
(NGFRAP1)(TNFRSF16) associated
protein 1
ASNADEnerve growth factor receptor3612986273548431787
(NGFRAP1)(TNFRSF16) associated
protein 1
ASNADEnerve growth factor receptor3726346283617171788
(NGFRAP1)(TNFRSF16) associated
protein 1
ASNADEnerve growth factor receptor3726356293617181789
(NGFRAP1)(TNFRSF16) associated
protein 1
ASNADEnerve growth factor receptor3726456303617281790
(NGFRAP1)(TNFRSF16) associated
protein 1
ASNGFnerve growth factor (beta3695126313585251791
polypeptide)
ASNGFRnerve growth factor receptor1722296321722291792
ASNGFRnerve growth factor receptor5042016334217311793
ASNIKmitogen-activated protein3446866343420591794
(MAP3K14)kinase kinase kinase 14
ASNIKmitogen-activated protein3769266353661251795
(MAP3K14)kinase kinase kinase 14
ASNOXAphorbol-12-myristate-13-2695186362695181796
acetate-induced protein 1
ASNOXAphorbol-12-myristate-13-3166606373261191797
acetate-induced protein 1
ASOX40tumor necrosis factor receptor3792366383685381798
superfamily, member 4
ASOX40tumor necrosis factor receptor4535806393909071799
superfamily, member 4
ASOX40Ltumor necrosis factor (ligand)2818346402818341800
(TNFSF4)superfamily, member 4
ASOX40Ltumor necrosis factor (ligand)3677186413566911801
TNFSF4)superfamily, member 4
ASOX40Ltumor necrosis factor (ligand)5452926424397041802
TNFSF4)superfamily, member 4
ASp53tumor protein p532693056432693051803
ASp53tumor protein p5326930564426930518042489
ASp53tumor protein p533595976453526101805
ASp53tumor protein p533964736463797351806
ASp53tumor protein p534134656474107391807
ASp53tumor protein p534143156483941951808
ASp53tumor protein p534190246494021301809
ASp53tumor protein p534202466503911271810
ASp53tumor protein p5344588865139147818112490
ASp53tumor protein p534552636523988461812
ASp53tumor protein p535035916534262521813
ASp53tumor protein p535087936544241041814
ASp53tumor protein p535096906554251041815
ASp53tumor protein p535149446564238621816
ASp53tumor protein p535458586574377921817
ASp70 S6ribosomal protein S6 kinase,2255776582255771818
kinase 170 kDa, polypeptide 1
ASp70 S6ribosomal protein S6 kinase,3930216593767441819
kinase 170 kDa, polypeptide 1
ASp70 S6ribosomal protein S6 kinase,4061166603843351820
kinase 170 kDa, polypeptide 1
ASp70 S6ribosomal protein S6 kinase,4435726614419931821
kinase 170 kDa, polypeptide 1
ASp70 S6ribosomal protein S6 kinase,3126296623084131822
kinase 270 kDa, polypeptide 2
ASp70 S6ribosomal protein S6 kinase,5289646634328471823
kinase 270 kDa, polypeptide 2
ASp70 S6ribosomal protein S6 kinase,5391886644429491824
kinase 270 kDa, polypeptide 2
ASp90Rskribosomal protein S6 kinase,3741626653632771825
90 kDa, polypeptide 1
ASp90Rskribosomal protein S6 kinase,3741646663632791826
90 kDa, polypeptide 1
ASp90Rskribosomal protein S6 kinase,3741686673632831827
90 kDa, polypeptide 1
ASp90Rskribosomal protein S6 kinase,4037326683839671828
90 kDa, polypeptide 1
ASp90Rskribosomal protein S6 kinase,5300036694322811829
90 kDa, polypeptide 1
ASp90Rskribosomal protein S6 kinase,5313826704354121830
90 kDa, polypeptide 1
ASPAK2p21 protein (Cdc42/Rac)-3271346713140671831
activated kinase 2
ASPARP-1poly (ADP-ribose) polymerase 13667906723557551832
ASPARP-1poly (ADP-ribose) polymerase 13667916733557561833
ASPARP-1poly (ADP-ribose) polymerase 13667926743557571834
ASPARP-1poly (ADP-ribose) polymerase 13667946753557591835
ASPARP-1poly (ADP-ribose) polymerase 14323386764127741836
ASPDPK13-phosphoinositide dependent3420856773442201837
protein kinase-1
ASPDPK13-phosphoinositide dependent3548366783468951838
protein kinase-1
ASPDPK13-phosphoinositide dependent4415496793953571839
protein kinase-1
ASPI3Kphosphoinositide-3-kinase,2639676802639671840
catalytic, alpha polypeptide
ASPI3Kphosphoinositide-3-kinase,2891536812891531841
catalytic, beta polypeptide
ASPI3Kphosphoinositide-3-kinase,3591956823521211842
catalytic, gamma polypeptide
ASPI3Kphosphoinositide-3-kinase,3605636833537661843
catalytic, delta polypeptide
ASPI3Kphosphoinositide-3-kinase,3611106843544101844
catalytic, delta polypeptide
ASPI3Kphosphoinositide-3-kinase,3773466853665631845
catalytic, delta polypeptide
ASPI3Kphosphoinositide-3-kinase,4406506863922581846
catalytic, gamma polypeptide
ASPI3Kphosphoinositide-3-kinase,4614516874203991847
catalytic, beta polypeptide
ASPI3Kphosphoinositide-3-kinase,4680366884174791848
catalytic, alpha polypeptide
ASPI3Kphosphoinositide-3-kinase,4775936894181431849
catalytic, beta polypeptide
ASPI3Kphosphoinositide-3-kinase,4839686904198571850
catalytic, beta polypeptide
ASPI3Kphosphoinositide-3-kinase,4935686914178691851
catalytic, beta polypeptide
ASPI3Kphosphoinositide-3-kinase,4961666924192601852
catalytic, gamma polypeptide
ASPI3Kphosphoinositide-3-kinase,5366566934464441853
catalytic, delta polypeptide
ASPI3Kphosphoinositide-3-kinase,5433906944438111854
catalytic, delta polypeptide
ASPI3Kphosphoinositide-3-kinase,5447166954382591855
catalytic, beta polypeptide
ASPKA-catprotein kinase, cAMP-3086776963095911856
dependent, catalytic, alpha
ASPKA-catprotein kinase, cAMP-3503566973409401857
dependent, catalytic, alpha
ASPKA-catprotein kinase, cAMP-3706796983597131858
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706806993597141859
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706817003597151860
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706827013597161861
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706847023597181862
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706857033597191863
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706887043597221864
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3706897053597231865
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3772767063664881866
dependent, catalytic, gamma
ASPKA-catprotein kinase, cAMP-3948387073783141867
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-3948397083783151868
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-4135387093971751869
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-4175307103993261870
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-4321117113922751871
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-4361337123909061872
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-4465387134012521873
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-4507307143936541874
dependent, catalytic, beta
ASPKA-catprotein kinase, cAMP-5356957154416541875
dependent, catalytic, alpha
ASPKA-catprotein kinase, cAMP-5366497164404181876
dependent, catalytic, alpha
ASPKC-protein kinase C, delta3304527173316021877
delta
ASPKC-protein kinase C, delta3947297183782171878
delta
ASPKC-protein kinase C, delta4788437194197261879
delta
ASPKC-protein kinase C, delta4878977204181061880
delta
ASPKC-protein kinase C, zeta3785677213678301881
Zeta
ASPKC-protein kinase C, zeta4009207223837111882
Zeta
ASPKC-protein kinase C, zeta4009217233837121883
Zeta
ASPKC-protein kinase C, zeta4611067244264121884
Zeta
ASPKC-protein kinase C, zeta4705117254213501885
Zeta
ASPKC-protein kinase C, zeta4705967264242281886
Zeta
ASPKC-protein kinase C, zeta4709867274212191887
Zeta
ASPKC-protein kinase C, zeta4826867284253171888
Zeta
ASPKC-protein kinase C, zeta4963257294218691889
Zeta
ASPP1-catprotein phosphatase 1, catalytic3129897303260311890
alphasubunit, alpha isozyme
ASPP1-catprotein phosphatase 1, catalytic3767457313659361891
alphasubunit, alpha isozyme
ASPP1-catprotein phosphatase 1, catalytic4514587324056031892
alphasubunit, alpha isozyme
ASPP2aprotein phosphatase 2, catalytic4811957334184471893
catalyticsubunit, alpha isozyme
ASPP2Cprotein phosphatase,2287057342287051894
Mg2+/Mn2+ dependent, 1H
ASPP2Cprotein phosphatase,2632127352632121895
Mg2+/Mn2+ dependent, 1F
ASPP2Cprotein phosphatase,2824127362824121896
Mg2+/Mn2+ dependent, 1B
ASPP2Cprotein phosphatase,2959087372959081897
Mg2+/Mn2+ dependent, 1K
ASPP2Cprotein phosphatase,2964877382964871898
Mg2+/Mn2+ dependent, 1M
ASPP2Cprotein phosphatase,3059217393066821899
Mg2+/Mn2+ dependent, 1D
ASPP2Cprotein phosphatase,3082497403124111900
Mg2+/Mn2+ dependent, 1E
ASPP2Cprotein phosphatase,3092767413089261901
Mg2+/Mn2+ dependent, 1J
ASPP2Cprotein phosphatase,3151947423247611902
Mg2+/Mn2+ dependent, 1K
ASPP2Cprotein phosphatase,3235887433198941903
Mg2+/Mn2+ dependent, 1M
ASPP2Cprotein phosphatase,3246887443217611904
Mg2+/Mn2+ dependent, 1N
(putative)
ASPP2Cprotein phosphatase,3256427453272551905
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,3256587463148501906
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,3440347473427781907
Mg2+/Mn2+ dependent, 1G
ASPP2Cprotein phosphatase,3452497483260891908
Mg2+/Mn2+ dependent, 1B
ASPP2Cprotein phosphatase,3508037492647141909
Mg2+/Mn2+ dependent, 1G
ASPP2Cprotein phosphatase,3599947503530881910
Mg2+/Mn2+ dependent, 1J
ASPP2Cprotein phosphatase,3785517513678131911
Mg2+/Mn2+ dependent, 1B
ASPP2Cprotein phosphatase,3929957523767201912
Mg2+/Mn2+ dependent, 1D
ASPP2Cprotein phosphatase,3950767533785141913
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,3955437543789131914
Mg2+/Mn2+ dependent, 1G
ASPP2Cprotein phosphatase,3967347553799601915
Mg2+/Mn2+ dependent, 1N
(putative)
ASPP2Cprotein phosphatase,3974957563806321916
Mg2+/Mn2+ dependent, 1F
ASPP2Cprotein phosphatase,4069817573847151917
Mg2+/Mn2+ dependent, 1F
ASPP2Cprotein phosphatase,4071427583849301918
Mg2+/Mn2+ dependent, 1F
ASPP2Cprotein phosphatase,4094327593872871919
Mg2+/Mn2+ dependent, 1B
ASPP2Cprotein phosphatase,4095027603870461920
Mg2+/Mn2+ dependent, 1M
ASPP2Cprotein phosphatase,4098957613873411921
Mg2+/Mn2+ dependent, 1B
ASPP2Cprotein phosphatase,4198077623900871922
Mg2+/Mn2+ dependent, 1B
ASPP2Cprotein phosphatase,4431217633902571923
Mg2+/Mn2+ dependent, 1E
ASPP2Cprotein phosphatase,4573517643937471924
Mg2+/Mn2+ dependent, 1M
ASPP2Cprotein phosphatase,4973437654203541925
Mg2+/Mn2+ dependent, 1L
ASPP2Cprotein phosphatase,4981657664176591926
Mg2+/Mn2+ dependent, 1L
ASPP2Cprotein phosphatase,5064237674241551927
Mg2+/Mn2+ dependent, 1K
ASPP2Cprotein phosphatase,5253997684353981928
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,5282417694314531929
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,5295747704329661930
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,5319377714355751931
Mg2+/Mn2+ dependent, 1A
ASPP2Cprotein phosphatase,5381917724399151932
Mg2+/Mn2+ dependent, 1F
ASPP2Cprotein phosphatase,5444127734425361933
Mg2+/Mn2+ dependent, 1G
ASPP2Cprotein phosphatase,5447127744385181934
Mg2+/Mn2+ dependent, 1D
ASPumaBCL2 binding component 33008807753008801935
ASPumaBCL2 binding component 33419837763411551936
ASPumaBCL2 binding component 34390967773958621937
ASPumaBCL2 binding component 34492287784045031938
ASRAIDDCASP2 and RIPK1 domain3328967793276471939
containing adaptor with death
domain
ASRAIDDCASP2 and RIPK1 domain5418137804426241940
containing adaptor with death
domain
ASRAIDDCASP2 and RIPK1 domain5428937814390681941
containing adaptor with death
domain
ASRAIDDCASP2 and RIPK1 domain5510657824484251942
containing adaptor with death
domain
ASRANKtumor necrosis factor receptor2694857832694851943
superfamily, member 11a,
NFKB activator
ASRANKtumor necrosis factor receptor3827907843722401944
superfamily, member 11a,
NFKB activator
ASRANKLtumor necrosis factor (ligand)2398497852398491945
superfamily, member 11
ASRANKLtumor necrosis factor (ligand)3585457863513471946
superfamily, member 11
ASRANKLtumor necrosis factor (ligand)3987957873817751947
superfamily, member 11
ASRANKLtumor necrosis factor (ligand)4052627883840421948
superfamily, member 11
ASRANKLtumor necrosis factor (ligand)5448627894449131949
superfamily, member 11
ASReIAv-rel reticuloendotheliosis viral3086397903115081950
(p65oncogene hom*olog A (avian)
NF-
kappaB
subunit)
ASReIAv-rel reticuloendotheliosis viral4062467913842731951
(p65oncogene hom*olog A (avian)
NF-
kappaB
subunit)
ASReIAv-rel reticuloendotheliosis viral4266177924379801952
(p65oncogene hom*olog A (avian)
NF-
kappaB
subunit)
ASReIAv-rel reticuloendotheliosis viral5256937934325371953
(p65oncogene hom*olog A (avian)
NF-
kappaB
subunit)
ASReIAv-rel reticuloendotheliosis viral5262837944352901954
(p65oncogene hom*olog A (avian)
NF-
kappaB
subunit)
ASReIAv-rel reticuloendotheliosis viral5458167954437001955
(p65oncogene hom*olog A (avian)
NF-
kappaB
subunit)
ASRIPK1receptor (TNFRSF)-interacting2598087962598081956
serine-threonine kinase 1
ASRIPK1receptor (TNFRSF)-interacting3804097973697731957
serine-threonine kinase 1
ASRIPK1receptor (TNFRSF)-interacting4534837984159811958
serine-threonine kinase 1
ASRIPK1receptor (TNFRSF)-interacting5417917994422941959
serine-threonine kinase 1
ASSequestosome 1sequestosome 13607188003539441960
(p62)
ASSequestosome 1sequestosome 13769298013661281961
(p62)
ASSequestosome 1sequestosome 13898058023744551962
(p62)
ASSequestosome 1sequestosome 14028748033855531963
(p62)
ASSequestosome 1sequestosome 14222458043945341964
(p62)
ASSequestosome 1sequestosome 14543788054081071965
(p62)
ASSequestosome 1sequestosome 15140938064273081966
(p62)
ASShcSHC (Src hom*ology 2 domain2645548072645541967
containing) transforming
protein 2
ASShcSHC (Src hom*ology 2 domain3664428083961621968
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3684418093574261969
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3684438103574281970
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3684458113574301971
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3684498123574341972
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3684508133574351973
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3684538143574381974
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain3758308153649901975
containing) transforming
protein 3
ASShcSHC (Src hom*ology 2 domain3758318163649911976
containing) transforming
protein 3
ASShcSHC (Src hom*ology 2 domain3758358173649951977
containing) transforming
protein 3
ASShcSHC (Src hom*ology 2 domain4121708183984411978
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain4141158194049081979
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain4441798203988641980
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain4446648213963331981
containing) transforming
protein 1
ASShcSHC (Src hom*ology 2 domain4481168224013031982
containing) transforming
protein 1
ASSiah-1seven in absentia hom*olog 13567218233491561983
(Drosophila)
ASSiah-1seven in absentia hom*olog 13800068243693431984
(Drosophila)
ASSiah-1seven in absentia hom*olog 13947258253782141985
(Drosophila)
ASSMACdiablo, IAP-bindingNA826NA1986
mitochondrial protein
ASSmac/Diablodiablo, IAP-binding2671698272671691987
mitochondrial protein
ASSmac/Diablodiablo, IAP-binding3535488283203431988
mitochondrial protein
ASSmac/Diablodiablo, IAP-binding4139188294116381989
mitochondrial protein
ASSmac/Diablodiablo, IAP-binding4436498303984951990
mitochondrial protein
ASSmac/Diablodiablo, IAP-binding4649428314423601991
mitochondrial protein
ASSODDBCL2-associated athanogene 42873228322873221992
ASSODDBCL2-associated athanogene 44324718333932981993
ASSOSson of sevenless hom*olog 22163738342163731994
(Drosophila)
ASSOSson of sevenless hom*olog 12638798352638791995
(Drosophila)
ASSOSson of sevenless hom*olog 13950388363784791996
(Drosophila)
ASSOSson of sevenless hom*olog 14022198373846751997
(Drosophila)
ASSOSson of sevenless hom*olog 14260168383877841998
(Drosophila)
ASSOSson of sevenless hom*olog 14287218393999921999
(Drosophila)
ASSOSson of sevenless hom*olog 25436808404453282000
(Drosophila)
ASSOSson of sevenless hom*olog 15436988414411722001
(Drosophila)
ASSUMO-1SMT3 suppressor of mif two 33922448423760752002
hom*olog 1 (S. cerevisiae)
ASSUMO-1SMT3 suppressor of mif two 33922458433760762003
hom*olog 1 (S. cerevisiae)
ASSUMO-1SMT3 suppressor of mif two 33922468443760772004
hom*olog 1 (S. cerevisiae)
ASSUMO-1SMT3 suppressor of mif two 34092058453862672005
hom*olog 1 (S. cerevisiae)
ASSUMO-1SMT3 suppressor of mif two 34094988463864722006
hom*olog 1 (S. cerevisiae)
ASSurvivinbaculoviral IAP repeat3016338473016332007
containing 5
ASSurvivinbaculoviral IAP repeat3500518483241802008
containing 5
ASSurvivinbaculoviral IAP repeat3749488493640862009
containing 5
ASSurvivinbaculoviral IAP repeat4320148503890882010
containing 5
ASTACItumor necrosis factor receptor2616528512616522011
superfamily, member 13B
ASTACItumor necrosis factor receptor4375388524134532012
superfamily, member 13B
AStBidtBIDNANA2013
ASTL1Atumor necrosis factor (ligand)3740448533631562014
superfamily, member 15
ASTL1Atumor necrosis factor (ligand)3740458543631572015
superfamily, member 15
ASTNF-tumor necrosis factor3761228553652902016
alpha
ASTNF-tumor necrosis factor3834968563729882017
alpha
ASTNF-tumor necrosis factor4122758573928582018
alpha
ASTNF-tumor necrosis factor4204258584106682019
alpha
ASTNF-tumor necrosis factor4437078593894922020
alpha
ASTNF-tumor necrosis factor4452328603892652021
alpha
ASTNF-tumor necrosis factor4487818613894902022
alpha
ASTNF-tumor necrosis factor4492648623986982023
alpha
ASTNF-R1tumor necrosis factor receptor1627498631627492024
superfamily, member 1A
ASTNF-R1tumor necrosis factor receptor3661598643803892025
superfamily, member 1A
ASTNF-R2tumor necrosis factor receptor3762598653654352026
superfamily, member 1B
ASTNF-R2tumor necrosis factor receptor37625986636543520272491
superfamily, member 1B
ASTNF-R2tumor necrosis factor receptor4008638673836602028
superfamily, member 1B
ASTNF-R2tumor necrosis factor receptor5367828684404252029
superfamily, member 1B
ASTRADDTNFRSF1A-associated via3450578693412682030
death domain
ASTRAF2TNF receptor-associated factor 22476688702476682031
ASTRAF2TNF receptor-associated factor 23596628713526852032
ASTRAF2TNF receptor-associated factor 23716458723607082033
ASTRAF2TNF receptor-associated factor 24145898733976532034
ASTRAF2TNF receptor-associated factor 24190578744058602035
ASTRAF2TNF receptor-associated factor 24295098754065242036
ASTRAF2TNF receptor-associated factor 24327858764000612037
ASTRAF2TNF receptor-associated factor 25364688774464142038
ASTRAF3TNF receptor-associated factor 33476628783280032039
ASTRAF3TNF receptor-associated factor 33516918793324682040
ASTRAF3TNF receptor-associated factor 33927458803765002041
ASTRAF3TNF receptor-associated factor 35397218814459982042
ASTRAF3TNF receptor-associated factor 35603718824542072043
ASTRAF3TNF receptor-associated factor 35604638834536232044
ASTRAF5TNF receptor-associated factor 52614648842614642045
ASTRAF5TNF receptor-associated factor 53361848853368252046
ASTRAF5TNF receptor-associated factor 53670048863559712047
ASTRAF5TNF receptor-associated factor 54279258873898912048
ASTRAF6TNF receptor-associated factor 63481248883378532049
ASTRAF6TNF receptor-associated factor 65269958894336232050
ASTrkAneurotrophic tyrosine kinase,3681968903571792051
receptor, type 1
ASTrkAneurotrophic tyrosine kinase,3923028913761202052
receptor, type 1
ASTrkAneurotrophic tyrosine kinase,5243778924314182053
receptor, type 1
ASTWEAKtumor necrosis factor (ligand)2938258932938252054
(TNFSF12)superfamily, member 12
ASTWEAKtumor necrosis factor (ligand)5572338944514512055
(TNFSF12)superfamily, member 12
ASVDAC 1voltage-dependent anion2653338952653332056
channel 1
ASVDAC 1voltage-dependent anion3950448963784842057
channel 1
ASVDAC 1voltage-dependent anion3950478973784872058
channel 1
ASVDAC 2voltage-dependent anion2984688982984682059
channel 2
ASVDAC 2voltage-dependent anion3131328993616352060
channel 2
ASVDAC 2voltage-dependent anion3322119003616862061
channel 2
ASVDAC 2voltage-dependent anion3440369013448762062
channel 2
ASVDAC 2voltage-dependent anion4132899023895512063
channel 2
ASVDAC 2voltage-dependent anion4476779034014922064
channel 2
ASVDAC 2voltage-dependent anion5355539044459012065
channel 2
ASVDAC 2voltage-dependent anion5433519054430922066
channel 2
ASXIAPX-linked inhibitor of apoptosis3556409063478582067
ASXIAPX-linked inhibitor of apoptosis3711999073602422068
ASXIAPX-linked inhibitor of apoptosis4306259084006372069
ASXIAPX-linked inhibitor of apoptosis4347539093952302070
ASXIAPX-linked inhibitor of apoptosisNA910NA2071
CC/SATMataxia telangiectasia mutated2786169112786162072
CC/SATMataxia telangiectasia mutated3895119123741622073
CC/SATMataxia telangiectasia mutated4525089133880582074
CC/SATMataxia telangiectasia mutated5329319144323182075
CC/SATRataxia telangiectasia and Rad33507219153437412076
related
CC/SATRataxia telangiectasia and Rad33831019163725812077
related
CC/SATRIPATR interacting protein3202119173230992078
CC/SATRIPATR interacting protein3466919183023382079
CC/SATRIPATR interacting protein3571059193496202080
CC/SATRIPATR interacting protein4120529204009302081
CC/SATRIPATR interacting protein4211759214066642082
CC/SBard1BRCA1 associated RING2609479222609472083
domain 1
CC/SBard1BRCA1 associated RING4499679234067522084
domain 1
CC/SBLMBloom syndrome, RecQ3551129243472322085
helicase-like
CC/SBLMBloom syndrome, RecQ5369259254423302086
helicase-like
CC/SBLMBloom syndrome, RecQ5439779264390752087
helicase-like
CC/SBrca1breast cancer 1, early onset3094869273109382088
CC/SBrca1breast cancer 1, early onset3463159282469072089
CC/SBrca1breast cancer 1, early onset3516669293380072090
CC/SBrca1breast cancer 1, early onset3529939303122362091
CC/SBrca1breast cancer 1, early onset3540719313260022092
CC/SBrca1breast cancer 1, early onset3576549323502832093
CC/SBrca1breast cancer 1, early onset3936919333772942094
CC/SBrca1breast cancer 1, early onset4120619343971452095
CC/SBrca1breast cancer 1, early onset4612219354185482096
CC/SBrca1breast cancer 1, early onset4617989364179882097
CC/SBrca1breast cancer 1, early onset4683009374171482098
CC/SBrca1breast cancer 1, early onset4700269384192742099
CC/SBrca1breast cancer 1, early onset4711819394189602100
CC/SBrca1breast cancer 1, early onset4767779404175542101
CC/SBrca1breast cancer 1, early onset4771529414199882102
CC/SBrca1breast cancer 1, early onset4785319424204122103
CC/SBrca1breast cancer 1, early onset4840879434194812104
CC/SBrca1breast cancer 1, early onset4890379444207812105
CC/SBrca1breast cancer 1, early onset4917479454207052106
CC/SBrca1breast cancer 1, early onset4928599464202532107
CC/SBrca1breast cancer 1, early onset4937959474187752108
CC/SBrca1breast cancer 1, early onset4939199484188192109
CC/SBrca1breast cancer 1, early onset4941239494191032110
CC/SBrca1breast cancer 1, early onset4974889504189862111
CC/Sc-Ablc-abl oncogene 1, non-receptor3185609513233152112
tyrosine kinase
CC/Sc-Ablc-abl oncogene 1, non-receptor3723489523614232113
tyrosine kinase
CC/Sc-Ablc-abl oncogene 1, non-receptor3932939533769712114
tyrosine kinase
CC/Sc-Ablc-abl oncogene 1, non-receptor4384269544077562115
tyrosine kinase
CC/Sc-Ablc-abl oncogene 1, non-receptor4449709554004122116
tyrosine kinase
CC/SCDC25Acell division cycle 25 hom*olog3025069563037062117
A (S. pombe)
CC/SCDC25Acell division cycle 25 hom*olog3512319573431662118
A (S. pombe)
CC/SCDC25Acell division cycle 25 hom*olog4379729584042852119
A (S. pombe)
CC/SCDC25Bcell division cycle 25 hom*olog2459609592459602120
B (S. pombe)
CC/SCDC25Bcell division cycle 25 hom*olog3408339603391702121
B (S. pombe)
CC/SCDC25Bcell division cycle 25 hom*olog3442569613391252122
B (S. pombe)
CC/SCDC25Bcell division cycle 25 hom*olog3795989623689182123
B (S. pombe)
CC/SCDC25Bcell division cycle 25 hom*olog4398809634059722124
B (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog3237609643216562125
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog3489839653452052126
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog3565059663488982127
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog3572749673498212128
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog4151309683926312129
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog5030229694272512130
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog5139709704247952131
C (S. pombe)
CC/SCDC25Ccell division cycle 25 hom*olog5348929714431962132
C (S. pombe)
CC/SCDK2cyclin-dependent kinase 22669709722669702133
CC/SCDK2cyclin-dependent kinase 23540569732430672134
CC/SCDK4cyclin-dependent kinase 42579049742579042135
CC/SCDK4cyclin-dependent kinase 43129909753168892136
CC/SCDK4cyclin-dependent kinase 45403259764390762137
CC/SCDK4cyclin-dependent kinase 45522549774491792138
CC/SCDK4cyclin-dependent kinase 45523889784489632139
CC/SCDK4cyclin-dependent kinase 45528629794467632140
CC/SCDK6cyclin-dependent kinase 62657349802657342141
CC/SCDK6cyclin-dependent kinase 64248489813970872142
CC/SChk1checkpoint kinase 12789169822789162143
CC/SChk1checkpoint kinase 14288309834125042144
CC/SChk1checkpoint kinase 14380159843886482145
CC/SChk1checkpoint kinase 15247379854328902146
CC/SChk1checkpoint kinase 15253969864341412147
CC/SChk1checkpoint kinase 15269379874318152148
CC/SChk1checkpoint kinase 15270139884315252149
CC/SChk1checkpoint kinase 15340709894353712150
CC/SChk1checkpoint kinase 15346859904324702151
CC/SChk1checkpoint kinase 15443739914423172152
CC/SChk2checkpoint kinase 23283549923291782153
CC/SChk2checkpoint kinase 23482959933290122154
CC/SChk2checkpoint kinase 23825639943720032155
CC/SChk2checkpoint kinase 23825659953720062156
CC/SChk2checkpoint kinase 23825669963720072157
CC/SChk2checkpoint kinase 23825789973720212158
CC/SChk2checkpoint kinase 23825809983720232159
CC/SChk2checkpoint kinase 24027319993848352160
CC/SChk2checkpoint kinase 240364210003849192161
CC/SChk2checkpoint kinase 240427610013857472162
CC/SChk2checkpoint kinase 240559810023860872163
CC/SChk2checkpoint kinase 254477210034424582164
CC/SClaspinclaspin25119510042511952165
CC/SClaspinclaspin31812110053129952166
CC/SClaspinclaspin37322010063623172167
CC/SClaspinclaspin54435610074423352168
CC/SCyclin Acyclin A227402610082740262169
CC/SCyclin Bcyclin B125644210092564422170
CC/SCyclin Bcyclin B327601410102760142171
CC/SCyclin Bcyclin B228820710112882072172
CC/SCyclin Bcyclin B334860310123386822173
CC/SCyclin Bcyclin B337603810133652062174
CC/SCyclin Bcyclin B337604210143652102175
CC/SCyclin Bcyclin B339654010153797902176
CC/SCyclin Bcyclin B150550010164245882177
CC/SCyclin Bcyclin B150657210174233872178
CC/SCyclin Dcyclin D122750710182275072179
CC/SCyclin Dcyclin D226125410192612542180
CC/SCyclin Dcyclin D337298710203620782181
CC/SCyclin Dcyclin D337298810213620792182
CC/SCyclin Dcyclin D337299110223620822183
CC/SCyclin Dcyclin D341420010233975452184
CC/SCyclin Dcyclin D341549710244015952185
CC/SCyclin Dcyclin D350506410254258302186
CC/SCyclin Dcyclin D351164210264262122187
CC/SCyclin Dcyclin D154289710274418632188
CC/SCyclin Ecyclin E126264310282626432189
CC/SCyclin Ecyclin E230810810293091812190
CC/SCyclin Ecyclin E135794310303506252191
CC/SCyclin Ecyclin E239613310313794372192
CC/SCyclin Ecyclin E144498310324101792193
CC/SCyclin Ecyclin E252050910334290892194
CC/SCyclin Ecyclin E254272510344457262195
CC/SDNA-protein kinase, DNA-activated,31419110353134202196
PKcatalytic polypeptide
CC/SDNA-protein kinase, DNA-activated,33836810363451822197
PKcatalytic polypeptide
CC/SE2F1/2/E2F transcription factor 5,25611710372561172198
3/4/5/6p130-binding
CC/SE2F1/2/E2F transcription factor 630723610383021592199
3/4/5/6
CC/SE2F1/2/E2F transcription factor 134338010393455712200
3/4/5/6
CC/SE2F1/2/E2F transcription factor 334661810402629042201
3/4/5/6
CC/SE2F1/2/E2F transcription factor 236172910413552492202
3/4/5/6
CC/SE2F1/2/E2F transcription factor 636200910423550362203
3/4/5/6
CC/SE2F1/2/E2F transcription factor 337864610433679142204
3/4/5/6
CC/SE2F1/2/E2F transcription factor 4,37937810443686862205
3/4/5/6p107/p130-binding
CC/SE2F1/2/E2F transcription factor 638152510453709362206
3/4/5/6
CC/SE2F1/2/E2F transcription factor 5,41627410463981242207
3/4/5/6p130-binding
CC/SE2F1/2/E2F transcription factor 5,41893010474143122208
3/4/5/6p130-binding
CC/SE2F1/2/E2F transcription factor 5,51747610484291202209
3/4/5/6p130-binding
CC/SE2F1/2/E2F transcription factor 5,51823410494296692210
3/4/5/6p130-binding
CC/SE2F1/2/E2F transcription factor 353543210504434182211
3/4/5/6
CC/SE2F1/2/E2F transcription factor 654210010514463152212
3/4/5/6
CC/SE2F1/2/E2F transcription factor 654621210524388642213
3/4/5/6
CC/SFANCD2Fanconi anemia,28764710532876472214
complementation group D2
CC/SFANCD2Fanconi anemia,38380610543733172215
complementation group D2
CC/SFANCD2Fanconi anemia,38380710553733182216
complementation group D2
CC/SFANCD2Fanconi anemia,41958510563987542217
complementation group D2
CC/SFANCLFanconi anemia,23374110572337412218
complementation group L
CC/SFANCLFanconi anemia,54064610584414312219
complementation group L
CC/SGADD4growth arrest and DNA-37098610593600252220
5 alphadamage-inducible, alpha
CC/SGADD4growth arrest and DNA-21563110602156312221
5 betadamage-inducible, beta
CC/SGADD4growth arrest and DNA-37098510613600242222
5 betadamage-inducible, alpha
CC/SMDM2Mdm2 p53 binding protein25814810622581482223
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein25814910632581492224
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein29925210642992522225
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein31142010653107422226
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein31144010663113022227
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein34880110673350962228
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein35005710682666242229
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein35629010693486372230
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein35848310703512702231
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein36043010713536112232
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein39341010723770622233
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein39341210733770642234
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein39341310743770652235
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein39341510753770672236
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein42886310764106942237
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein46228410774172812238
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein51785210784302572239
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein53947910794444302240
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein54082710804409322241
hom*olog (mouse)
CC/SMDM2Mdm2 p53 binding protein54464810814432742242
hom*olog (mouse)
CC/SNFBD1mediator of DNA-damage37640510823655872243
checkpoint 1
CC/SNFBD1mediator of DNA-damage37640610833655882244
checkpoint 1
CC/SNFBD1mediator of DNA-damage38356610843730602245
checkpoint 1
CC/SNFBD1mediator of DNA-damage41239510853928332246
checkpoint 1
CC/SNFBD1mediator of DNA-damage41397310864088312247
checkpoint 1
CC/SNFBD1mediator of DNA-damage41636810874103832248
checkpoint 1
CC/SNFBD1mediator of DNA-damage41657110884009792249
checkpoint 1
CC/SNFBD1mediator of DNA-damage41703310894089622250
checkpoint 1
CC/SNFBD1mediator of DNA-damage41722810904003052251
checkpoint 1
CC/SNFBD1mediator of DNA-damage41917210913984742252
checkpoint 1
CC/SNFBD1mediator of DNA-damage41967510923976422253
checkpoint 1
CC/SNFBD1mediator of DNA-damage42001910933964842254
checkpoint 1
CC/SNFBD1mediator of DNA-damage42032010944165112255
checkpoint 1
CC/SNFBD1mediator of DNA-damage42210410953903752256
checkpoint 1
CC/SNFBD1mediator of DNA-damage42219510964077032257
checkpoint 1
CC/SNFBD1mediator of DNA-damage42226610974113102258
checkpoint 1
CC/SNFBD1mediator of DNA-damage42372610983912302259
checkpoint 1
CC/SNFBD1mediator of DNA-damage42443710993981512260
checkpoint 1
CC/SNFBD1mediator of DNA-damage42450711003883552261
checkpoint 1
CC/SNFBD1mediator of DNA-damage42463811013940742262
checkpoint 1
CC/SNFBD1mediator of DNA-damage42502911023971262263
checkpoint 1
CC/SNFBD1mediator of DNA-damage42507211033969892264
checkpoint 1
CC/SNFBD1mediator of DNA-damage42579011043970212265
checkpoint 1
CC/SNFBD1mediator of DNA-damage42740611053874292266
checkpoint 1
CC/SNFBD1mediator of DNA-damage42961011064068502267
checkpoint 1
CC/SNFBD1mediator of DNA-damage43035811074141632268
checkpoint 1
CC/SNFBD1mediator of DNA-damage43144111083927842269
checkpoint 1
CC/SNFBD1mediator of DNA-damage43299811094059912270
checkpoint 1
CC/SNFBD1mediator of DNA-damage43566411104043182271
checkpoint 1
CC/SNFBD1mediator of DNA-damage43579711114006772272
checkpoint 1
CC/SNFBD1mediator of DNA-damage43775911123877432273
checkpoint 1
CC/SNFBD1mediator of DNA-damage43816511133877062274
checkpoint 1
CC/SNFBD1mediator of DNA-damage44036911144152122275
checkpoint 1
CC/SNFBD1mediator of DNA-damage44139711153904892276
checkpoint 1
CC/SNFBD1mediator of DNA-damage44441211164136102277
checkpoint 1
CC/SNFBD1mediator of DNA-damage44513011173961242278
checkpoint 1
CC/SNFBD1mediator of DNA-damage44576411183938862279
checkpoint 1
CC/SNFBD1mediator of DNA-damage44719211194058062280
checkpoint 1
CC/SNFBD1mediator of DNA-damage44764011203963892281
checkpoint 1
CC/SNFBD1mediator of DNA-damage44889511213961212282
checkpoint 1
CC/SNFBD1mediator of DNA-damage44915311224091672283
checkpoint 1
CC/SNFBD1mediator of DNA-damage45003311233900402284
checkpoint 1
CC/SNFBD1mediator of DNA-damage45221311244049362285
checkpoint 1
CC/SNFBD1mediator of DNA-damage45572911254049542286
checkpoint 1
CC/SNFBD1mediator of DNA-damage45658911264053502287
checkpoint 1
CC/SNFBD1mediator of DNA-damage54648711274486792288
checkpoint 1
CC/SNFBD1mediator of DNA-damage54653911284482322289
checkpoint 1
CC/SNFBD1mediator of DNA-damage54704711294490592290
checkpoint 1
CC/SNFBD1mediator of DNA-damage54735311304478832291
checkpoint 1
CC/SNFBD1mediator of DNA-damage54768111314478512292
checkpoint 1
CC/SNFBD1mediator of DNA-damage54770011324490832293
checkpoint 1
CC/SNFBD1mediator of DNA-damage54787411334476822294
checkpoint 1
CC/SNFBD1mediator of DNA-damage54810311344494992295
checkpoint 1
CC/SNFBD1mediator of DNA-damage54811211354484342296
checkpoint 1
CC/SNFBD1mediator of DNA-damage54824811364480802297
checkpoint 1
CC/SNFBD1mediator of DNA-damage54843311374499712298
checkpoint 1
CC/SNFBD1mediator of DNA-damage54854211384465972299
checkpoint 1
CC/SNFBD1mediator of DNA-damage54880511394469242300
checkpoint 1
CC/SNFBD1mediator of DNA-damage54882711404492012301
checkpoint 1
CC/SNFBD1mediator of DNA-damage54889311414479432302
checkpoint 1
CC/SNFBD1mediator of DNA-damage54894711424477112303
checkpoint 1
CC/SNFBD1mediator of DNA-damage54922811434475172304
checkpoint 1
CC/SNFBD1mediator of DNA-damage54938211444491772305
checkpoint 1
CC/SNFBD1mediator of DNA-damage54942811454470382306
checkpoint 1
CC/SNFBD1mediator of DNA-damage54977111464488122307
checkpoint 1
CC/SNFBD1mediator of DNA-damage55000411474470842308
checkpoint 1
CC/SNFBD1mediator of DNA-damage55011011484469802309
checkpoint 1
CC/SNFBD1mediator of DNA-damage55021011494476972310
checkpoint 1
CC/SNFBD1mediator of DNA-damage55040811504471362311
checkpoint 1
CC/SNFBD1mediator of DNA-damage55050011514500022312
checkpoint 1
CC/SNFBD1mediator of DNA-damage55068811524480662313
checkpoint 1
CC/SNFBD1mediator of DNA-damage55120411534477992314
checkpoint 1
CC/SNFBD1mediator of DNA-damage55126711544501982315
checkpoint 1
CC/SNFBD1mediator of DNA-damage55146011554492742316
checkpoint 1
CC/SNFBD1mediator of DNA-damage55155411564485382317
checkpoint 1
CC/SNFBD1mediator of DNA-damage55162111574482852318
checkpoint 1
CC/SNFBD1mediator of DNA-damage55174011584500372319
checkpoint 1
CC/SNFBD1mediator of DNA-damage55226311594470692320
checkpoint 1
CC/SNFBD1mediator of DNA-damage55234911604498922321
checkpoint 1
CC/SNFBD1mediator of DNA-damage55247411614477712322
checkpoint 1
CC/SNFBD1mediator of DNA-damage55252211624499362323
checkpoint 1
CC/SNFBD1mediator of DNA-damage55277611634478252324
checkpoint 1
CC/SNFBD1mediator of DNA-damage55304711644472472325
checkpoint 1
CC/SNFBD1mediator of DNA-damage55304811654477872326
checkpoint 1
CC/SNFBD1mediator of DNA-damage55313011664468092327
checkpoint 1
CC/SNFBD1mediator of DNA-damage55319611674495862328
checkpoint 1
CC/SNibrinnibrin26543311682654332329
CC/SNibrinnibrin45238711694452132330
CC/Sp107retinoblastoma-like 1 (p107)34435911703436462331
CC/Sp107retinoblastoma-like 1 (p107)37366411713627682332
CC/Sp130retinoblastoma-like 2 (p130)26213311722621332333
CC/Sp130retinoblastoma-like 2 (p130)37993511733692672334
CC/Sp130retinoblastoma-like 2 (p130)54440511744437442335
CC/Sp130retinoblastoma-like 2 (p130)54454511754446852336
CC/Sp21P21NA1176NA2337
CC/SPCNAproliferating cell nuclear37914311773684382338
antigen
CC/SPCNAproliferating cell nuclear37916011783684582339
antigen
CC/SRAD9RAD9 hom*olog A (S. pombe)30798011793113602340
CC/SRbretinoblastoma 126716311802671632341
protein
CC/SRbretinoblastoma 146750511814347022342
protein
CC/SRbretinoblastoma 154291711824376422343
protein
CC/SSMC1structural maintenance of32221311833234212344
chromosomes 1A
CC/SSMC1structural maintenance of34021311843449062345
chromosomes 1A
CC/SSMC1structural maintenance of37534011853644892346
chromosomes 1A
CC/SSMC1structural maintenance of42801411864135092347
chromosomes 1A
CC/SUSP1ubiquitin specific peptidase 133995011873435262348
CC/SUSP1ubiquitin specific peptidase 137114611883601882349
CC/SUSP1ubiquitin specific peptidase 145214311894036622350
M4EBP-1eukaryotic translation initiation33882511903406912351
factor 4E binding protein 1
MARNTaryl hydrocarbon receptor35439611913463722352
nuclear translocator
MARNTaryl hydrocarbon receptor35859511923514072353
nuclear translocator
MARNTaryl hydrocarbon receptor36897511933579712354
nuclear translocator
MARNTaryl hydrocarbon receptor39470011943781902355
nuclear translocator
MARNTaryl hydrocarbon receptor47184411954258992356
nuclear translocator
MARNTaryl hydrocarbon receptor50575511964275712357
nuclear translocator
MARNTaryl hydrocarbon receptor51519211974238512358
nuclear translocator
MCAIXcarbonic anhydrase IX37835711983676082359
MCAIXcarbonic anhydrase IX54407411994385412360
MCBPCREB binding protein26236712002623672361
MCBPCREB binding protein32350812013235502362
MCBPCREB binding protein38207012023715022363
MCBPCREB binding protein54388312034419782364
MCITED1Cbp/p300-interacting24613912042461392365
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting37361912053627212366
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting41740012064147812367
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting42741212073914072368
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting42979412084074962369
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting43138112093885482370
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting44598312104032742371
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting45087512114057652372
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED1Cbp/p300-interacting45370712124017642373
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 1
MCITED2Cbp/p300-interacting36765112133566232374
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 2
MCITED2Cbp/p300-interacting39231212143761262375
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 2
MCITED2Cbp/p300-interacting53615912154428312376
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 2
MCITED2Cbp/p300-interacting53733212164441982377
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 2
MCITED4Cbp/p300-interactingNA1217NA2378
transactivator, with Glu/Asp-
rich carboxy-terminal domain, 4
MCITED4Cbp/p300-interacting37263812183617212379
transactivator, with Glu/Asp-
rich carboxy-terminal domain,
4 (CBP/p300 interacting
transactivator with ED-rich
tail)
MCOMMD1copper metabolism (Murr1)31183212193082362380
domain containing 1
MCOMMD1copper metabolism (Murr1)42741712204132072381
domain containing 1
MCOMMD1copper metabolism (Murr1)44416612214100502382
domain containing 1
MCOMMD1copper metabolism (Murr1)45833712224012362383
domain containing 1
MCOMMD1copper metabolism (Murr1)53873612234389612384
domain containing 1
MCREBcAMP responsive element23699612242369962385
binding protein 1
MCREBcAMP responsive element35326712252369952386
binding protein 1
MCREBcAMP responsive element35370412263421362387
binding protein 3
MCREBcAMP responsive element37439712273635182388
binding protein 1
MCREBcAMP responsive element43062412284055392389
binding protein 1
MCREBcAMP responsive element43232912293876992390
binding protein 1
MCREBcAMP responsive element44580312304072272391
binding protein 1
MCREBcAMP responsive element45247412313924282392
binding protein 1
MCREBcAMP responsive element53672612324458922393
binding protein 1
MCREBcAMP responsive element53978912334408092394
binding protein 1
MeIF4Eeukaryotic translation initiation28089212342808922395
factor 4E
MeIF4Eeukaryotic translation initiation45025312353896242396
factor 4E
MHIF3-hypoxia inducible factor 3,24430212362443022397
alphaalpha subunit
MHIF3-hypoxia inducible factor 3,29130012372913002398
alphaalpha subunit
MFIHhypoxia inducible factor 1,29916312382991632399
alpha subunit inhibitor (factor
inhibiting HIF)
MHIF3-hypoxia inducible factor 3,30086212393008622400
alpha)alpha subunit
MHIF3-hypoxia inducible factor 3,33961312403418772401
alphaalpha subunit
MHIF3-hypoxia inducible factor 3,37767012413668982402
alphaalpha subunit
MHIF3-hypoxia inducible factor 3,41470712424128082403
alphaalpha subunit
MHIF3-hypoxia inducible factor 3,42010212434077712404
alphaalpha subunit
MFIHhypoxia inducible factor 1,44272412443997342405
(factoralpha subunit inhibitor
inhibiting
HIF)
MHIF3-hypoxia inducible factor 3,45777112454080082406
alphaalpha subunit
MHIF3-hypoxia inducible factor 3,45786512463940522407
alphaalpha subunit
MHIF3-hypoxia inducible factor 3,47543212474325782408
alphaalpha subunit
MFIHhypoxia inducible factor 1,53358912484333602409
(factoralpha subunit inhibitor
inhibiting
HIF)
MGrb2growth factor receptor-bound31661512493173602410
protein 2
MGrb2growth factor receptor-bound31680412503390072411
protein 2
MGrb2growth factor receptor-bound39256212513763452412
protein 2
MGrb2growth factor receptor-bound39256412523763472413
protein 2
MHNF4alphahepatocyte nuclear factor 4,31609912533129872414
alpha
MHNF4alphahepatocyte nuclear factor 4,31667312543151802415
alpha
MHNF4alphahepatocyte nuclear factor 4,33869212553438072416
alpha
MHNF4alphahepatocyte nuclear factor 4,41569112564121112417
alpha
MHNF4alphahepatocyte nuclear factor 4,44359812574109112418
alpha
MHNF4alphahepatocyte nuclear factor 4,45723212583962162419
alpha
MHNF4alpha2hom*o sapiens hepatocyteNA1259NA2420
nuclear factor 4, alpha
(HNF4A), transcript variant 2,
mRNA
MIBP3insulin-like growth factor27552112602755212421
binding protein 3
MIBP3insulin-like growth factor38108312613704732422
binding protein 3
MIBP3insulin-like growth factor38108612623704762423
binding protein 3
MIBP3insulin-like growth factor41762112633991162424
binding protein 3
MIBP3insulin-like growth factor42853012643902982425
binding protein 3
MIBP3insulin-like growth factor43304712654044612426
binding protein 3
MIBP3insulin-like growth factor43849112663937402427
binding protein 3
MIBP3insulin-like growth factor44214212673924722428
binding protein 3
MIBP3insulin-like growth factor54503212684399992429
binding protein 3
MJAB1COP9 constitutive35784912693505122430
photomorphogenic hom*olog
subunit 5 (Arabidopsis)
MMNK1MAP kinase interacting34118312703395732431
serine/threonine kinase 1
MMNK1MAP kinase interacting37194412713610122432
serine/threonine kinase 1
MMNK1MAP kinase interacting37194512723610132433
serine/threonine kinase 1
MMNK1MAP kinase interacting37194612733610142434
serine/threonine kinase 1
MMNK1MAP kinase interacting42811212744111352435
serine/threonine kinase 1
MMNK1MAP kinase interacting49661912754367092436
serine/threonine kinase 1
MMNK1MAP kinase interacting54573012764409742437
serine/threonine kinase 1
MMNK2MAP kinase interacting25089612772508962438
serine/threonine kinase 2
MMNK2MAP kinase interacting30934012783094852439
serine/threonine kinase 2
MMNK2MAP kinase interacting54116512794389042440
serine/threonine kinase 2
MMNK2MAP kinase interacting54562712804412452441
serine/threonine kinase 2
Mp15(INK4A)cyclin-dependent kinase27692512812769252442
inhibitor 2B (p15, inhibits
CDK4)
Mp15(INK4A)cyclin-dependent kinase38014212823694872443
inhibitor 2B (p15, inhibits
CDK4)
Mp300E1A binding protein p30026325312832632532444
MPer1period hom*olog 1 (Drosophila)31727612843144202445
MPer1period hom*olog 1 (Drosophila)35490312853469792446
MRPS6ribosomal protein S631537712863697432447
MRPS6ribosomal protein S638038112873697412448
MRPS6ribosomal protein S638038412883697452449
MRPS6ribosomal protein S638039412893697572450
MSHARP1basic helix-loop-helix family,NA1290NA2451
member e41
MSHARP1basic helix-loop-helix family,24272812912427282452
(BHLHE41)member e41
MSHARP1basic helix-loop-helix family,54073112924373692453
(BHLHE41)member e41
MSRC1nuclear receptor coactivator 128859912932885992454
MSRC1nuclear receptor coactivator 134833212943209402455
MSRC1nuclear receptor coactivator 139585612953791972456
MSRC1nuclear receptor coactivator 140514112963850972457
MSRC1nuclear receptor coactivator 140696112973852162458
MSRC1nuclear receptor coactivator 153853912984440392459
Mtuberintuberous sclerosis 221947612992194762460
Mtuberintuberous sclerosis 235077313003443832461
Mtuberintuberous sclerosis 235392913012480992462
Mtuberintuberous sclerosis 238253813023719782463
Mtuberintuberous sclerosis 240187413033844682464
Mtuberintuberous sclerosis 243967313043992322465
AIFSHapoptosis-inducing factor,NA1305NA2466
short
Angiopoietin1Angiopoietin 1NA1306NA24672492
BMP2BMP2 CONA1307NA24682493
CO
c-MYCv-myc myelocytomatosis viralNA1308NA2469
oncogene hom*olog (avian)
COMMD1COMMD1NA1309NA
COMMD1COMMD1 with nuclear exportNANA2470
NESseqences deleted
deleted
COMMD1COMMD1 with nuclear exportNANA2471
NES1sequences deleted and nuclear
deletedlocalization signals added
and NLS
added
COMMD1COMMD1 with SV40 andNANA2472
SV40nuclear localization signals
NLS
COMMD1wtCOMMD1 wild-typeNANA2473
GLUT1solute carrier family 2NA1310NA2474
(facilitated glucose
transporter), member 1
GranulysinGranulysin FL15NA1311NA2475
FL15
GranulysinGranulysin NS9NANA24762494
NS9
GranulysinGranulysin S9NANA24772495
S9
HIF1 ahypoxia inducible factor 1,NA1312NA2478
alpha subunit (basic helix-
loop-helix transcription factor)
IL15interleukin 15NA1313NA2479
KGFfibroblast growth factor 7,NA1314NA2480
precursor; mature is 32-194
MCT4solute carrier family 16,NA1315NA24812496
member 4 (monocarboxylic
acid transporter 5)
MYCMYC inhibitor D (OMOMyc)NA1316NA2482
inhibitor D
MYCMYC inhibitor D_90NANA2483
inhibitor(OmoMyc_90)
D_90
C.A.Constitutively active (C.A.)NA1317NA2484
caspasecaspase 3 cleavable
3_cleavable(RevCasp3_Cleavable)
C.A.Constitutively active (C.A.)NA1318NA2485
caspasecaspase 3 uncleavable
3_uncleavable(RevCasp3_UnCleavable)
C.A.Constitutively active (C.A.)NA1319NA2486
caspase 6caspase 6 (RevCasp6)
SIAh1siah E3 ubiquitin protein ligase 1NA1320NA2487
HSV1-tkHerpes simplex virus 1-
thymidine kinase

Shown in Table 7, are familiar cancer syndromes, tumor suppressor genes, function of the tumor suppressor gene, chromosomal location, and tumor type observed. Signal-sensor polynucleotides of the present invention can be designed as a therapeutic for any of those listed in the table.

TABLE 7
Familial Cancer Syndrome Targets
FamilialTumor
CancerSuppressorChromosomalTumor Types
SyndromeGeneFunctionLocationObserved
Li-FraumeniP53cell cycle17p13.1brain tumors,
Syndromeregulation,sarcomas, leukemia,
apoptosisbreast cancer
FamilialRB1cell cycle13q14.1-q14.2retinoblastoma,
Retinoblastomaregulationosteogenic sarcoma
Wilms TumorWT1transcriptional11p13pediatric kidney
regulationcancer, most
common form of
childhood solid
tumor
NeurofibromatosisNF1catalysis of RAS17q11.2neurofibromas,
Type 1inactivationsarcomas, gliomas
NeurofibromatosisNF2linkage of cell22q12.2Schwann cell
Type 2membrane to actintumors,
cytoskeletonastrocytomas,
meningiomas,
ependymonas
FamilialAPCsignaling through5q21-q22colon cancer
Adenomatousadhesion
Polyposismolecules to
nucleus
TuberousTSC1forms complex9q34seizures, mental
sclerosis 1with TSC2retardation, facial
protein, inhibitsangiofibromas
signaling to
downstream
effectors of mTOR
TuberousTSC2forms complex16p13.3benign growths
sclerosis 2with TSC1(hamartomas) in
protein, inhibitsmany tissues,
signaling toastrocytomas,
downstreamrhabdomyosarcomas
effectors of mTOR
Deleted inDPC4, alsoregulation of18q21.1pancreatic
Pancreaticknown asTGF-β/BMPcarcinoma, colon
Carcinoma 4,SMAD4signal transductioncancer
Familial
juvenile
polyposis
syndrome
Deleted inDCCtransmembrane18q21.3colorectal cancer
Colorectalreceptor involved
Carcinomain axonal guidance
via netrins
Familial BreastBRCA1functions in17q21breast and ovarian
Cancertranscription,cancer
DNA binding,
transcription
coupled DNA
repair,
hom*ologous
recombination,
chromosomal
stability,
ubiquitination of
proteins, and
centrosome
replication
Familial BreastBRCA2transcriptional13q12.3breast and ovarian
Cancer(FANCD1)regulation ofcancer
genes involved in
DNA repair and
hom*ologous
recombination
CowdenPTENphosphoinositide10q23.3gliomas, breast
syndrome3-phosphatase,cancer, thyroid
protein tyrosinecancer, head & neck
phosphatasesquamous carcinoma
Peutz-JeghersSTK11phosphorylates19p13.3hyperpigmentation,
Syndrome (PJS)(serine-and activatesmultiple
threonineAMP-activatedhamartomatous
kinase 11)kinase (AMPK),polyps, colorectal,
AMPK involvedbreast and ovarian
in stresscancers
responses, lipid
and glucose
meatabolism
HereditaryMSH2DNA mismatch2p22-p21colon cancer
Nonpolyposisrepair
Colon Cancer
type 1,
HNPCC1
HereditaryMLH1DNA mismatch3p21.3colon cancer
Nonpolyposisrepair
Colon Cancer
type 2,
HNPCC2
Familial diffuse-CDH1cell-cell adhesion16q22.1gastric cancer,
type gastricproteinlobular breast cancer
cancer
von Hippel-VHLregulation of3p26-p25renal cancers,
Lindautranscriptionhemangioblastomas,
Syndromeelongation throughpheochromocytoma,
activation of aretinal angioma
ubiquitin ligase
complex
FamilialCDKN2Ap16INK49p21melanoma,
Melanomainhibits cell-cyclepancreatic cancer,
kinases CDK4 andothers
CDK6; p14ARF
binds
the p53 stabilizing
protein MDM2
GorlinPTCHtransmembrane9q22.3basal cell skin
Syndrome:(e.g.,receptor for soniccarcinoma
Nevoid basalPTCH1,hedgehog (shh),
cell carcinomaPTCH2)involved in early
syndromedevelopment
(NBCCS)through repression
of action of
smoothened
MultipleMEN1intrastrand DNA11q13parathyroid and
Endocrinecrosslink repairpituitary adenomas,
Neoplasia Type 1islet cell tumors,
carcinoid

In additional to the above mentioned targets, the oncology-related polypeptides may include any “death signal” protein that can be recognized by active T cells of immune system. Such suicide signal proteins encoded by the sensor-signal polynucleotides can be selectively expressed in particular tissues or cells (e.g. cancer cells) through engineered microRNA binding sites and/or other regulatory elements as described herein. The group of proteins, when they are expressed on the surface of a caner cell, can prime T cell to induce T cell mediated immune response, thus killing the cancer cell. As a non-limiting example, a group of proteins that are known to present a “death signal”, include, CD80, CD86, B7 and MHC II, etc.

Protein Cleavage Signals and Sites

In one embodiment, the oncology-related polypeptides of the present invention may include at least one protein cleavage signal containing at least one protein cleavage site. The protein cleavage site may be located at the N-terminus, the C-terminus, at any space between the N- and the C-termini such as, but not limited to, half-way between the N- and C-termini, between the N-terminus and the half way point, between the half way point and the C-terminus, and combinations thereof.

The oncology-related polypeptides of the present invention may include, but is not limited to, a proprotein convertase (or prohormone convertase), thrombin or Factor Xa protein cleavage signal. Proprotein convertases are a family of nine proteinases, comprising seven basic amino acid-specific subtilisin-like serine proteinases related to yeast kexin, known as prohormone convertase 1/3 (PC1/3), PC2, furin, PC4, PC5/6, paired basic amino-acid cleaving enzyme 4 (PACE4) and PC7, and two other subtilases that cleave at non-basic residues, called subtilisin kexin isozyme 1 (SKI-1) and proprotein convertase subtilisin kexin 9 (PCSK9). Non-limiting examples of protein cleavage signal amino acid sequences are listing in Table 8. In Table 8, “X” refers to any amino acid, “n” may be 0, 2, 4 or 6 amino acids and “*” refers to the protein cleavage site. In Table 8, SEQ ID NO: 2499 refers to when n=4 and SEQ ID NO: 2500 refers to when n=6.

TABLE 8
Protein Cleavage Site Sequences
Protein Cleavage
SignalAmino Acid Cleavage SequenceSEQ ID NO
ProproteinR-X-X-R*2497
convertaseR-X-K/R-R*2498
K/R-Xn-K/R*2499 or 2500
ThrombinL-V-P-R*-G-S2501
L-V-P-R*2502
A/F/G/I/L/T/V/M-A/F/G/I/L/T/V/W/A-2503
P-R*
Factor XaI-E-G-R*2504
I-D-G-R*2505
A-E-G-R*2506
A/F/G/I/L/T/V/M-D/E-G-R*2507

In one embodiment, the signal-sensor primary constructs and the mmRNA of the present invention may be engineered such that the primary construct or mmRNA contains at least one encoded protein cleavage signal. The encoded protein cleavage signal may be located before the start codon, after the start codon, before the coding region, within the coding region such as, but not limited to, half way in the coding region, between the start codon and the half way point, between the half way point and the stop codon, after the coding region, before the stop codon, between two stop codons, after the stop codon and combinations thereof.

In one embodiment, the signal-sensor primary constructs or mmRNA of the present invention may include at least one encoded protein cleavage signal containing at least one protein cleavage site. The encoded protein cleavage signal may include, but is not limited to, a proprotein convertase (or prohormone convertase), thrombin and/or Factor Xa protein cleavage signal. One of skill in the art may use Table 1 above or other known methods to determine the appropriate encoded protein cleavage signal to include in the signal-sensor primary constructs or mmRNA of the present invention. For example, starting with the signal of Table 8 and considering the codons of Table 1 one can design a signal for the signal-sensor primary construct which can produce a protein signal in the resulting oncology-related polypeptide.

In one embodiment, the oncology-related polypeptides of the present invention include at least one protein cleavage signal and/or site.

As a non-limiting example, U.S. Pat. No. 7,374,930 and U.S. Pub. No. 20090227660, herein incorporated by reference in their entireties, use a furin cleavage site to cleave the N-terminal methionine of GLP-1 in the expression product from the Golgi apparatus of the cells. In one embodiment, the polypeptides of the present invention include at least one protein cleavage signal and/or site with the proviso that the polypeptide is not GLP-1.

In one embodiment, the signal-sensor primary constructs or mmRNA of the present invention includes at least one encoded protein cleavage signal and/or site.

In one embodiment, the signal-sensor primary constructs or mmRNA of the present invention includes at least one encoded protein cleavage signal and/or site with the proviso that the signal-sensor primary construct or mmRNA does not encode GLP-1.

In one embodiment, the signal-sensor primary constructs or mmRNA of the present invention may include more than one coding region. Where multiple coding regions are present in the signal-sensor primary construct or mmRNA of the present invention, the multiple coding regions may be separated by encoded protein cleavage sites. As a non-limiting example, the signal-sensor primary construct or mmRNA may be signed in an ordered pattern. On such pattern follows AXBY form where A and B are coding regions which may be the same or different coding regions and/or may encode the same or different oncology-related polypeptides, and X and Y are encoded protein cleavage signals which may encode the same or different protein cleavage signals. A second such pattern follows the form AXYBZ where A and B are coding regions which may be the same or different coding regions and/or may encode the same or different oncology-related polypeptides, and X, Y and Z are encoded protein cleavage signals which may encode the same or different protein cleavage signals. A third pattern follows the form ABXCY where A, B and C are coding regions which may be the same or different coding regions and/or may encode the same or different oncology-related polypeptides, and X and Y are encoded protein cleavage signals which may encode the same or different protein cleavage signals.

In one embodiment, the oncology-related polypeptides, signal-sensor primary constructs and mmRNA can also contain sequences that encode protein cleavage sites so that the polypeptides, signal-sensor primary constructs and mmRNA can be released from a carrier region or a fusion partner by treatment with a specific protease for said protein cleavage site.

microRNA

microRNAs (or miRNA) are 19-25 nucleotide long noncoding RNAs that bind to the 3′UTR of nucleic acid molecules and down-regulate gene expression either by reducing nucleic acid molecule stability or by inhibiting translation. The modified nucleic acids (mRNA), enhanced modified RNA or ribonucleic acids of the invention may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds. Such sequences may correspond to any known microRNA such as those taught in US Publication US2005/0261218 and US Publication US2005/0059005, the contents of which are incorporated herein by reference in their entirety. As a non-limiting embodiment, known microRNAs, their sequences and their binding site sequences in the human genome are listed below in Table 9.

A microRNA sequence comprises a “seed” region, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick complementarity to the miRNA target sequence. A microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA. In some embodiments, a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1. In some embodiments, a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1. See for example, Grimson A, Farh K K, Johnston W K, Garrett-Engele P, Lim L P, Bartel D P; Mol Cell. 2007 Jul. 6; 27(1):91-105. The bases of the microRNA seed have complete complementarity with the target sequence. By engineering microRNA target sequences into the 3′UTR of nucleic acids or mRNA of the invention one can target the molecule for degradation or reduced translation, provided the microRNA in question is available. This process will reduce the hazard of off target effects upon nucleic acid molecule delivery. Identification of microRNA, microRNA target regions, and their expression patterns and role in biology have been reported (Bonauer et al., Curr Drug Targets 2010 11:943-949; Anand and Cheresh Curr Opin Hematol 2011 18:171-176; Contreras and Rao Leukemia 2012 26:404-413 (2011 Dec. 20. doi: 10.1038/leu.2011.356); Bartel Cell 2009 136:215-233; Landgraf et al, Cell, 2007 129:1401-1414; Gentner and Naldini, Tissue Antigens. 2012 80:393-403 and all references therein; each of which is herein incorporated by reference in its entirety).

For example, if the signal-sensor polynucleotide is not intended to be delivered to the liver but ends up there, then miR-122, a microRNA abundant in liver, can inhibit the expression of the gene of interest if one or multiple target sites of miR-122 are engineered into the 3′UTR of the signal-sensor polynucleotide. Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation of a signal-sensor polynucleotide. As used herein, the term “microRNA site” refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that “binding” may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.

Conversely, for the purposes of the signal-sensor polynucleotides of the present invention, microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues. For example, miR-122 binding sites may be removed to improve protein expression in the liver.

In one embodiment, signal-sensor polynucleotides may include at least one miRNA-binding site in the 3′UTR in order to direct cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but not limited to, normal and/or cancerous cells (e.g., HEP3B or SNU449). As a non-limiting example, a strong apoptotic signal and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR. As another non-limiting example, apoptosis inducing factor short isoform (AIFsh) and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR. As yet another non-limiting example, constitutively active (C.A.) caspase 6 and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR. As another non-limiting example, HSV1-tk and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR.

In another embodiment, signal-sensor polynucleotides may include three miRNA-binding sites in the 3′UTR in order to direct cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but not limited to, normal and/or cancerous cells (e.g., HEP3B or SNU449). As a non-limiting example, a strong apoptotic signal and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR. As another non-limiting example, apoptosis inducing factor short isoform (AIFsh) and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR. As yet another non-limiting example, constitutively active (C.A.) caspase 6 and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR. As another non-limiting example, HSV1-tk and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR.

Regulation of expression in multiple tissues can be accomplished through introduction or removal or one or several microRNA binding sites. Shown below in Table 10 are microRNAs which are differentially expressed in different tissues and cells, and often associated with different types of diseases (e.g. cancer cells). The decision of removal or insertion of microRNA binding sites, or any combination, is dependent on microRNA expression patterns and their profilings in cancer cells.

Examples of tissues where microRNA are known to regulate mRNA, and thereby protein expression, include, but are not limited to, liver (miR-122), muscle (miR-133, miR-206, miR-208), endothelial cells (miR-17-92, miR-126), myeloid cells (miR-142-3p, miR-142-5p, miR-16, miR-21, miR-223, miR-24, miR-27), nervous system (mir-124a, miR-9), pluripotent cells (miR-302, miR-367, miR-290, miR-371, miR-373), pancreatic islet cells (miR-375), adipose tissue (let-7, miR-30c), heart (miR-1d, miR-149), kidney (miR-192, miR-194, miR-204), and lung epithelial cells (let-7, miR-133, miR-126).

Specifically, microRNAs are known to be differentially expressed in immune cells (also called hematopoietic cells), such as antigen presenting cells (APCs) (e.g. dendritic cells and macrophages), macrophages, monocytes, B lymphocytes, T lymphocytes, granulocytes, natural killer cells, etc. Immune cell specific microRNAs are involved in immunogenicity, autoimmunity, the immune-response to infection, inflammation, as well as unwanted immune response after gene therapy and tissue/organ transplantation. Immune cells specific microRNAs also regulate many aspects of development, proliferation, differentiation and apoptosis of hematopoietic cells (immune cells). For example, miR-142 and miR-146 are exclusively expressed in the immune cells, particularly abundant in myeloid dendritic cells. Introducing the miR-142 binding site into the 3′-UTR of a signal-sensor polypeptide of the present invention can selectively suppress the gene expression in the antigen presenting cells through miR-142 mediated mRNA degradation, limiting antigen presentation in professional APCs (e.g. dendritic cells) and thereby preventing antigen-mediated immune response after gene delivery (see, Annoni A et al., blood, 2009, 114, 5152-5161, the content of which is herein incorporated by reference in its entirety.)

In one embodiment, microRNAs binding sites that are known to be expressed in immune cells, in particular, the antigen presenting cells, can be engineered into the signal-sensor polynucleotides to suppress the expression of the sensor-signal polynucleotide in APCs through microRNA mediated RNA degradation, subduing the antigen-mediated immune response, while the expression of the sensor-signal polynucleotide is maintained in non-immune cells where the immune cell specific microRNAs are not expressed. For example, to prevent the immunogenic reaction caused by a liver specific protein expression, the miR-122 binding site can be removed and the miR-142 (and/or mirR-146) binding sites can be engineered into the 3-UTR of the signal-sensor polynucleotide (e.g., see the constructs described in Example 38 and the experiment outlined in Examples 39 and 40).

To further drive the selective degradation and suppression of mRNA in APCs and macrophage, the signal-sensor polynucleotide may include another negative regulatory element in the 3-UTR, either alone or in combination with mir-142 and/or mir-146 binding sites. As a non-limiting example, one regulatory element is the Constitutive Decay Elements (CDEs).

Immune cells specific microRNAs include, but are not limited to, hsa-let-7a-2-3p, hsa-let-7a-3p, hsa-7a-5p, hsa-let-7c, hsa-let-7e-3p, hsa-let-7e-5p, hsa-let-7g-3p, hsa-let-7g-5p, hsa-let-7i-3p, hsa-let-7i-5p, miR-10a-3p, miR-10a-5p, miR-1184, hsa-let-7f-1-3p, hsa-let-7f-2-5p, hsa-let-7f-5p, miR-125b-1-3p, miR-125b-2-3p, miR-125b-5p, miR-1279, miR-130a-3p, miR-130a-5p, miR-132-3p, miR-132-5p, miR-142-3p, miR-142-5p, miR-143-3p, miR-143-5p, miR-146a-3p, miR-146a-5p, miR-146b-3p, miR-146b-5p, miR-147a, miR-147b, miR-148a-5p, miR-148a-3p, miR-150-3p, miR-150-5p, miR-151b, miR-155-3p, miR-155-5p, miR-15a-3p, miR-15a-5p, miR-15b-5p, miR-15b-3p, miR-16-1-3p, miR-16-2-3p, miR-16-5p, miR-17-5p, miR-181a-3p, miR-181a-5p, miR-181a-2-3p, miR-182-3p, miR-182-5p, miR-197-3p, miR-197-5p, miR-21-5p, miR-21-3p, miR-214-3p, miR-214-5p, miR-223-3p, miR-223-5p, miR-221-3p, miR-221-5p, miR-23b-3p, miR-23b-5p, miR-24-1-5p, miR-24-2-5p, miR-24-3p, miR-26a-1-3p, miR-26a-2-3p, miR-26a-5p, miR-26b-3p, miR-26b-5p, miR-27a-3p, miR-27a-5p, miR-27b-3p, miR-27b-5p, miR-28-3p, miR-28-5p, miR-2909, miR-29a-3p, miR-29a-5p, miR-29b-1-5p, miR-29b-2-5p, miR-29c-3p, miR-29c-5p, miR-30e-3p, miR-30e-5p, miR-331-5p, miR-339-3p, miR-339-5p, miR-345-3p, miR-345-5p, miR-346, miR-34a-3p, miR-34a-5p, miR-363-3p, miR-363-5p, miR-372, miR-377-3p, miR-377-5p, miR-493-3p, miR-493-5p, miR-542, miR-548b-5p, miR548c-5p, miR-548i, miR-548j, miR-548n, miR-574-3p, miR-598, miR-718, miR-935, miR-99a-3p, miR-99a-5p, miR-99b-3p and miR-99b-5p. Shown below in Table 11 are microRNAs that are enriched in specific types of immune cells. Furthermore, novel miroRNAs are discovered in the immune cells in the art through micro-array hybridization and microtome analysis (Jima D D et al, Blood, 2010, 116:e118-e127; Vaz C et al., BMC Genomics, 2010, 11,288, the content of each of which is incorporated herein by reference in its entirety).

MicroRNAs that are known to be expressed in the liver include, but are not limited to, miR-107, miR-122-3p, miR-122-5p, miR-1228-3p, miR-1228-5p, miR-1249, miR-129-5p, miR-1303, miR-151a-3p, miR-151a-5p, miR-152, miR-194-3p, miR-194-5p, miR-199a-3p, miR-199a-5p, miR-199b-3p, miR-199b-5p, miR-296-5p, miR-557, miR-581, miR-939-3p, miR-939-5p. microRNA binding sites from any liver specific microRNA can be introduced to or removed from the signal-sensor polynucleotides to regulate the expression of the signal-sensor polynucleotides in the liver. Liver specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent an immune reaction against protein expression in the liver.

MicroRNAs that are known to be expressed in the lung include, but are not limited to, let-7a-2-3p, let-7a-3p, let-7a-5p, miR-126-3p, miR-126-5p, miR-127-3p, miR-127-5p, miR-130a-3p, miR-130a-5p, miR-130b-3p, miR-130b-5p, miR-133a, miR-133b, miR-134, miR-18a-3p, miR-18a-5p, miR-18b-3p, miR-18b-5p, miR-24-1-5p, miR-24-2-5p, miR-24-3p, miR-296-3p, miR-296-5p, miR-32-3p, miR-337-3p, miR-337-5p, miR-381-3p, miR-381-5p. MicroRNA binding sites from any lung specific microRNA can be introduced to or removed from the signal-sensor polynucleotide to regulate the expression of the signal-sensor polynucleotide in the lung. Lung specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent an immune reaction against protein expression in the lung.

MicroRNAs that are known to be expressed in the heart include, but are not limited to, miR-1, miR-133a, miR-133b, miR-149-3p, miR-149-5p, miR-186-3p, miR-186-5p, miR-208a, miR-208b, miR-210, miR-296-3p, miR-320, miR-451a, miR-451b, miR-499a-3p, miR-499a-5p, miR-499b-3p, miR-499b-5p, miR-744-3p, miR-744-5p, miR-92b-3p and miR-92b-5p. microRNA binding sites from any heart specific microRNA can be introduced to or removed from the signal-sensor polynucleotides to regulate the expression of the signal-sensor polynucleotides in the heart. Heart specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent an immune reaction against protein expression in the heart.

MicroRNAs that are known to be expressed in the nervous system include, but are not limited to, miR-124-5p, miR-125a-3p, miR-125a-5p, miR-125b-1-3p, miR-125b-2-3p, miR-125b-5p, miR-1271-3p, miR-1271-5p, miR-128, miR-132-5p, miR-135a-3p, miR-135a-5p, miR-135b-3p, miR-135b-5p, miR-137, miR-139-5p, miR-139-3p, miR-149-3p, miR-149-5p, miR-153, miR-181c-3p, miR-181c-5p, miR-183-3p, miR-183-5p, miR-190a, miR-190b, miR-212-3p, miR-212-5p, miR-219-1-3p, miR-219-2-3p, miR-23a-3p, miR-23a-5p, miR-30a-5p, miR-30b-3p, miR-30b-5p, miR-30c-1-3p, miR-30c-2-3p, miR-30c-5p, miR-30d-3p, miR-30d-5p, miR-329, miR-342-3p, miR-3665, miR-3666, miR-380-3p, miR-380-5p, miR-383, miR-410, miR-425-3p, miR-425-5p, miR-454-3p, miR-454-5p, miR-483, miR-510, miR-516a-3p, miR-548b-5p, miR-548c-5p, miR-571, miR-7-1-3p, miR-7-2-3p, miR-7-5p, miR-802, miR-922, miR-9-3p and miR-9-5p. microRNAs enriched in the nervous system further include those specifically expressed in neurons, including, but not limited to, miR-132-3p, miR-132-3p, miR-148b-3p, miR-148b-5p, miR-151a-3p, miR-151a-5p, miR-212-3p, miR-212-5p, miR-320b, miR-320e, miR-323a-3p, miR-323a-5p, miR-324-5p, miR-325, miR-326, miR-328, miR-922 and those specifically expressed in glial cells, including, but not limited to, miR-1250, miR-219-1-3p, miR-219-2-3p, miR-219-5p, miR-23a-3p, miR-23a-5p, miR-3065-3p, miR-3065-5p, miR-30e-3p, miR-30e-5p, miR-32-5p, miR-338-5p, miR-657. microRNA binding sites from any CNS specific microRNA can be introduced to or removed from the signal-sensor polynucleotides to regulate the expression of the signal-sensor polynucleotide in the nervous system. Nervous system specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent an immune reaction against protein expression in the nervous system.

MicroRNAs that are known to be expressed in the pancreas include, but are not limited to, miR-105-3p, miR-105-5p, miR-184, miR-195-3p, miR-195-5p, miR-196a-3p, miR-196a-5p, miR-214-3p, miR-214-5p, miR-216a-3p, miR-216a-5p, miR-30a-3p, miR-33a-3p, miR-33a-5p, miR-375, miR-7-1-3p, miR-7-2-3p, miR-493-3p, miR-493-5p and miR-944. MicroRNA binding sites from any pancreas specific microRNA can be introduced to or removed from the signal-sensor polynucleotide to regulate the expression of the signal-sensor polynucleotide in the pancreas. Pancreas specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent immune reaction against protein expression in the pancreas.

MicroRNAs that are known to be expressed in the kidney further include, but are not limited to, miR-122-3p, miR-145-5p, miR-17-5p, miR-192-3p, miR-192-5p, miR-194-3p, miR-194-5p, miR-20a-3p, miR-20a-5p, miR-204-3p, miR-204-5p, miR-210, miR-216a-3p, miR-216a-5p, miR-296-3p, miR-30a-3p, miR-30a-5p, miR-30b-3p, miR-30b-5p, miR-30c-1-3p, miR-30c-2-3p, miR30c-5p, miR-324-3p, miR-335-3p, miR-335-5p, miR-363-3p, miR-363-5p and miR-562. MicroRNA binding sites from any kidney specific microRNA can be introduced to or removed from the signal-sensor polynucleotide to regulate the expression of the signal-sensor polynucleotide in the kidney. Kidney specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent immune reaction against protein expression in the kidney.

MicroRNAs that are known to be expressed in the muscle further include, but are not limited to, let-7g-3p, let-7g-5p, miR-1, miR-1286, miR-133a, miR-133b, miR-140-3p, miR-143-3p, miR-143-5p, miR-145-3p, miR-145-5p, miR-188-3p, miR-188-5p, miR-206, miR-208a, miR-208b, miR-25-3p and miR-25-5p. MicroRNA binding sites from any muscle specific microRNA can be introduced to or removed from the signal-sensor polynucleotide to regulate the expression of the signal-sensor polynucleotide in the muscle. Muscle specific microRNAs binding sites can be engineered alone or further in combination with immune cells (e.g. APCs) microRNA binding sites in order to prevent an immune reaction against protein expression in the muscle.

MicroRNAs are differentially expressed in different types of cells, such as endothelial cells, epithelial cells and adipocytes. For example, microRNAs that are expressed in endothelial cells include, but are not limited to, let-7b-3p, let-7b-5p, miR-100-3p, miR-100-5p, miR-101-3p, miR-101-5p, miR-126-3p, miR-126-5p, miR-1236-3p, miR-1236-5p, miR-130a-3p, miR-130a-5p, miR-17-5p, miR-17-3p, miR-18a-3p, miR-18a-5p, miR-19a-3p, miR-19a-5p, miR-19b-1-5p, miR-19b-2-5p, miR-19b-3p, miR-20a-3p, miR-20a-5p, miR-217, miR-210, miR-21-3p, miR-21-5p, miR-221-3p, miR-221-5p, miR-222-3p, miR-222-5p, miR-23a-3p, miR-23a-5p, miR-296-5p, miR-361-3p, miR-361-5p, miR-421, miR-424-3p, miR-424-5p, miR-513a-5p, miR-92a-1-5p, miR-92a-2-5p, miR-92a-3p, miR-92b-3p and miR-92b-5p. Many novel microRNAs were discovered in endothelial cells from deep-sequencing analysis (Voellenkle C et al., RNA, 2012, 18, 472-484, herein incorporated by reference in its entirety). MicroRNA binding sites from any endothelial cell specific microRNA can be introduced to or removed from the signal-sensor polynucleotide in order to modulate the expression of the signal-sensor polynucleotide in the endothelial cells in various conditions.

For further example, microRNAs that are expressed in epithelial cells include, but are not limited to, let-7b-3p, let-7b-5p, miR-1246, miR-200a-3p, miR-200a-5p, miR-200b-3p, miR-200b-5p, miR-200c-3p, miR-200c-5p, miR-338-3p, miR-429, miR-451a, miR-451b, miR-494, miR-802 and miR-34a, miR-34b-5p, miR-34c-5p, miR-449a, miR-449b-3p, miR-449b-5p specific in respiratory ciliated epithelial cells; let-7 family, miR-133a, miR-133b, miR-126 specific in lung epithelial cells; miR-382-3p, miR-382-5p specific in renal epithelial cells and miR-762 specific in corneal epithelial cells. MicroRNA binding sites from any epithelial cell specific microRNA can be introduced to or removed from the signal-sensor polynucleotide in order to modulate the expression of the signal-sensor polynucleotide in the epithelial cells in various conditions.

In addition, a large group of microRNAs are enriched in embryonic stem cells, controlling stem cell self-renewal as well as the development and/or differentiation of various cell lineages, such as neural cells, cardiac, hematopoietic cells, skin cells, osteogenic cells and muscle cells (Kuppusamy K T et al., Curr. Mol Med, 2013, 13(5), 757-764; Vidigal J A and Ventura A, Semin Cancer Biol. 2012, 22(5-6), 428-436; Goff L A et al., PLoS One, 2009, 4:e7192; Morin R D et al., Genome Res, 2008, 18, 610-621; Yoo J K et al., Stem Cells Dev. 2012, 21(11), 2049-2057, each of which is herein incorporated by reference in its entirety). MicroRNAs abundant in embryonic stem cells include, but are not limited to, let-7a-2-3p, let-a-3p, let-7a-5p, let7d-3p, let-7d-5p, miR-103a-2-3p, miR-103a-5p, miR-106b-3p, miR-106b-5p, miR-1246, miR-1275, miR-138-1-3p, miR-138-2-3p, miR-138-5p, miR-154-3p, miR-154-5p, miR-200c-3p, miR-200c-5p, miR-290, miR-301a-3p, miR-301a-5p, miR-302a-3p, miR-302a-5p, miR-302b-3p, miR-302b-5p, miR-302c-3p, miR-302c-5p, miR-302d-3p, miR-302d-5p, miR-302e, miR-367-3p, miR-367-5p, miR-369-3p, miR-369-5p, miR-370, miR-371, miR-373, miR-380-5p, miR-423-3p, miR-423-5p, miR-486-5p, miR-520c-3p, miR-548e, miR-548f, miR-548g-3p, miR-548g-5p, miR-548i, miR-548k, miR-548l, miR-548m, miR-548n, miR-548o-3p, miR-548o-5p, miR-548p, miR-664a-3p, miR-664a-5p, miR-664b-3p, miR-664b-5p, miR-766-3p, miR-766-5p, miR-885-3p, miR-885-5p, miR-93-3p, miR-93-5p, miR-941, miR-96-3p, miR-96-5p, miR-99b-3p and miR-99b-5p. Many predicted novel microRNAs are discovered by deep sequencing in human embryonic stem cells (Morin R D et al., Genome Res, 2008, 18, 610-621; Goff L A et al., PLoS One, 2009, 4:e7192; Bar M et al., Stem cells, 2008, 26, 2496-2505, the content of each of which is incorporated herein by references in its entirety).

In one embodiment, the binding sites of embryonic stem cell specific microRNAs can be included in or removed from the 3-UTR of the signal-sensor polynucleotide to modulate the development and/or differentiation of embryonic stem cells, to inhibit the senescence of stem cells in a degenerative condition (e.g. degenerative diseases), or to stimulate the senescence and apoptosis of stem cells in a disease condition (e.g. cancer stem cell).

Many microRNA expression studies have been conducted, and are described in the art, to profile the differential expression of microRNAs in various cancer cells/tissues and other diseases. Some microRNAs are abnormally over-expressed in certain cancer cells and others are under-expressed. For example, microRNAs are differentially expressed in cancer cells (WO2008/154098, US2013/0059015, US2013/0042333, WO2011/157294); cancer stem cells (US2012/0053224); pancreatic cancers and diseases (US2009/0131348, US2011/0171646, US2010/0286232, U.S. Pat. No. 8,389,210); asthma and inflammation (U.S. Pat. No. 8,415,096); prostate cancer (US2013/0053264); hepatocellular carcinoma (WO2012/151212, US2012/0329672, WO2008/054828, U.S. Pat. No. 8,252,538); lung cancer cells (WO2011/076143, WO2013/033640, WO2009/070653, US2010/0323357); cutaneous T cell lymphoma (WO2013/011378); colorectal cancer cells (WO2011/0281756, WO2011/076142); cancer positive lymph nodes (WO2009/100430, US2009/0263803); nasopharyngeal carcinoma (EP2112235); chronic obstructive pulmonary disease (US2012/0264626, US2013/0053263); thyroid cancer (WO2013/066678); ovarian cancer cells (US2012/0309645, WO2011/095623); breast cancer cells (WO2008/154098, WO2007/081740, US2012/0214699), leukemia and lymphoma (WO2008/073915, US2009/0092974, US2012/0316081, US2012/0283310, WO2010/018563, the content of each of which is incorporated herein by reference in their entirety).

Specifically, microRNA sites that are over-expressed in certain cancer and/or tumor cells can be removed from the 3-UTR of the signal-sensor polynucleotide encoding the oncology-related polypeptide, restoring the expression suppressed by the over-expressed microRNAs in cancer cells, thus ameliorating the corresponsive biological function, for instance, transcription stimulation and/or repression, cell cycle arrest, apoptosis and cell death. Normal cells and tissues, wherein microRNA expression is not up-regulated, will remain unaffected.

MicroRNA can also regulate complex biological processes such as angiogenesis (miR-132) (Anand and Cheresh Curr Opin Hematol 2011 18:171-176). In the signal-sensor polynucleotides of the invention, binding sites for microRNAs that are involved in such processes may be removed or introduced, in order to tailor the expression of the signal-sensor polynucleotides expression to biologically relevant cell types or to the context of relevant biological processes. In this context, the signal-sensor polynucleotide are defined as auxotrophic signal-sensor polynucleotides.

Table 9 is a non-exhaustive listing of miRs and miR binding sites (miR BS) and their sequences which may be used with the present invention.

TABLE 9
Mirs and mir binding sites
mir SEQBS SEQ
microRNAIDID
hsa-let-7a-2-3p25083529
hsa-let-7a-3p25093530
hsa-let-7a-5p25103531
hsa-let-7b-3p25113532
hsa-let-7b-5p25123533
hsa-let-7c25133534
hsa-let-7d-3p25143535
hsa-let-7d-5p25153536
hsa-let-7e-3p25163537
hsa-let-7e-5p25173538
hsa-let-7f-1-3p25183539
hsa-let-7f-2-3p25193540
hsa-let-7f-5p25203541
hsa-let-7g-3p25213542
hsa-let-7g-5p25223543
hsa-let-7i-3p25233544
hsa-let-7i-5p25243545
hsa-miR-125253546
hsa-miR-100-3p25263547
hsa-miR-100-5p25273548
hsa-miR-101-3p25283549
hsa-miR-101-5p25293550
hsa-miR-103a-2-5p25303551
hsa-miR-103a-3p25313552
hsa-miR-103b25323553
hsa-miR-105-3p25333554
hsa-miR-105-5p25343555
hsa-miR-106a-3p25353556
hsa-miR-106a-5p25363557
hsa-miR-106b-3p25373558
hsa-miR-106b-5p25383559
hsa-miR-10725393560
hsa-miR-10a-3p25403561
hsa-miR-10a-5p25413562
hsa-miR-10b-3p25423563
hsa-miR-10b-5p25433564
hsa-miR-1178-3p25443565
hsa-miR-1178-5p25453566
hsa-miR-117925463567
hsa-miR-118025473568
hsa-miR-118125483569
hsa-miR-118225493570
hsa-miR-118325503571
hsa-miR-118425513572
hsa-miR-1185-1-3p25523573
hsa-miR-1185-2-3p25533574
hsa-miR-1185-5p25543575
hsa-miR-119325553576
hsa-miR-119725563577
hsa-miR-120025573578
hsa-miR-120225583579
hsa-miR-120325593580
hsa-miR-120425603581
hsa-miR-120525613582
hsa-miR-120625623583
hsa-miR-1207-3p25633584
hsa-miR-1207-5p25643585
hsa-miR-120825653586
hsa-miR-122-3p25663587
hsa-miR-1224-3p25673588
hsa-miR-1224-5p25683589
hsa-miR-1225-3p25693590
hsa-miR-1225-5p25703591
hsa-miR-122-5p25713592
hsa-miR-1226-3p25723593
hsa-miR-1226-5p25733594
hsa-miR-1227-3p25743595
hsa-miR-1227-5p25753596
hsa-miR-1228-3p25763597
hsa-miR-1228-5p25773598
hsa-miR-1229-3p25783599
hsa-miR-1229-5p25793600
hsa-miR-123125803601
hsa-miR-1233-1-5p25813602
hsa-miR-1233-3p25823603
hsa-miR-1234-3p25833604
hsa-miR-1234-5p25843605
hsa-miR-1236-3p25853606
hsa-miR-1236-5p25863607
hsa-miR-1237-3p25873608
hsa-miR-1237-5p25883609
hsa-miR-1238-3p25893610
hsa-miR-1238-5p25903611
hsa-miR-124325913612
hsa-miR-124-3p25923613
hsa-miR-124425933614
hsa-miR-1245a25943615
hsa-miR-1245b-3p25953616
hsa-miR-1245b-5p25963617
hsa-miR-124-5p25973618
hsa-miR-124625983619
hsa-miR-1247-3p25993620
hsa-miR-1247-5p26003621
hsa-miR-124826013622
hsa-miR-124926023623
hsa-miR-125026033624
hsa-miR-125126043625
hsa-miR-125226053626
hsa-miR-125326063627
hsa-miR-125426073628
hsa-miR-1255a26083629
hsa-miR-1255b-2-3p26093630
hsa-miR-1255b-5p26103631
hsa-miR-125626113632
hsa-miR-125726123633
hsa-miR-125826133634
hsa-miR-125a-3p26143635
hsa-miR-125a-5p26153636
hsa-miR-125b-1-3p26163637
hsa-miR-125b-2-3p26173638
hsa-miR-125b-5p26183639
hsa-miR-1260a26193640
hsa-miR-1260b26203641
hsa-miR-126126213642
hsa-miR-126226223643
hsa-miR-126326233644
hsa-miR-126-3p26243645
hsa-miR-126426253646
hsa-miR-126526263647
hsa-miR-126-5p26273648
hsa-miR-126626283649
hsa-miR-126726293650
hsa-miR-1268a26303651
hsa-miR-1268b26313652
hsa-miR-1269a26323653
hsa-miR-1269b26333654
hsa-miR-127026343655
hsa-miR-1271-3p26353656
hsa-miR-1271-5p26363657
hsa-miR-127226373658
hsa-miR-1273a26383659
hsa-miR-1273c26393660
hsa-miR-1273d26403661
hsa-miR-1273e26413662
hsa-miR-1273f26423663
hsa-miR-1273g-3p26433664
hsa-miR-1273g-5p26443665
hsa-miR-127-3p26453666
hsa-miR-127526463667
hsa-miR-127-5p26473668
hsa-miR-127626483669
hsa-miR-1277-3p26493670
hsa-miR-1277-5p26503671
hsa-miR-127826513672
hsa-miR-127926523673
hsa-miR-12826533674
hsa-miR-128126543675
hsa-miR-128226553676
hsa-miR-128326563677
hsa-miR-128426573678
hsa-miR-1285-3p26583679
hsa-miR-1285-5p26593680
hsa-miR-128626603681
hsa-miR-128726613682
hsa-miR-128826623683
hsa-miR-128926633684
hsa-miR-129026643685
hsa-miR-129126653686
hsa-miR-129-1-3p26663687
hsa-miR-1292-3p26673688
hsa-miR-129-2-3p26683689
hsa-miR-1292-5p26693690
hsa-miR-129326703691
hsa-miR-129426713692
hsa-miR-1295a26723693
hsa-miR-1295b-3p26733694
hsa-miR-1295b-5p26743695
hsa-miR-129-5p26753696
hsa-miR-129626763697
hsa-miR-129726773698
hsa-miR-129826783699
hsa-miR-129926793700
hsa-miR-130126803701
hsa-miR-130226813702
hsa-miR-130326823703
hsa-miR-1304-3p26833704
hsa-miR-1304-5p26843705
hsa-miR-130526853706
hsa-miR-1306-3p26863707
hsa-miR-1306-5p26873708
hsa-miR-1307-3p26883709
hsa-miR-1307-5p26893710
hsa-miR-130a-3p26903711
hsa-miR-130a-5p26913712
hsa-miR-130b-3p26923713
hsa-miR-130b-5p26933714
hsa-miR-132126943715
hsa-miR-132226953716
hsa-miR-132326963717
hsa-miR-132-3p26973718
hsa-miR-132426983719
hsa-miR-132-5p26993720
hsa-miR-133a27003721
hsa-miR-133b27013722
hsa-miR-13427023723
hsa-miR-134327033724
hsa-miR-135a-3p27043725
hsa-miR-135a-5p27053726
hsa-miR-135b-3p27063727
hsa-miR-135b-5p27073728
hsa-miR-136-3p27083729
hsa-miR-136-5p27093730
hsa-miR-13727103731
hsa-miR-138-1-3p27113732
hsa-miR-138-2-3p27123733
hsa-miR-138-5p27133734
hsa-miR-139-3p27143735
hsa-miR-139-5p27153736
hsa-miR-140-3p27163737
hsa-miR-140-5p27173738
hsa-miR-141-3p27183739
hsa-miR-141-5p27193740
hsa-miR-142-3p27203741
hsa-miR-142-5p27213742
hsa-miR-143-3p27223743
hsa-miR-143-5p27233744
hsa-miR-144-3p27243745
hsa-miR-144-5p27253746
hsa-miR-145-3p27263747
hsa-miR-145-5p27273748
hsa-miR-146827283749
hsa-miR-146927293750
hsa-miR-146a-3p27303751
hsa-miR-146a-5p27313752
hsa-miR-146b-3p27323753
hsa-miR-146b-5p27333754
hsa-miR-147027343755
hsa-miR-147127353756
hsa-miR-147a27363757
hsa-miR-147b27373758
hsa-miR-148a-3p27383759
hsa-miR-148a-5p27393760
hsa-miR-148b-3p27403761
hsa-miR-148b-5p27413762
hsa-miR-149-3p27423763
hsa-miR-149-5p27433764
hsa-miR-150-3p27443765
hsa-miR-150-5p27453766
hsa-miR-151a-3p27463767
hsa-miR-151a-5p27473768
hsa-miR-151b27483769
hsa-miR-15227493770
hsa-miR-15327503771
hsa-miR-153727513772
hsa-miR-153827523773
hsa-miR-153927533774
hsa-miR-154-3p27543775
hsa-miR-154-5p27553776
hsa-miR-155-3p27563777
hsa-miR-155-5p27573778
hsa-miR-158727583779
hsa-miR-15a-3p27593780
hsa-miR-15a-5p27603781
hsa-miR-15b-3p27613782
hsa-miR-15b-5p27623783
hsa-miR-16-1-3p27633784
hsa-miR-16-2-3p27643785
hsa-miR-16-5p27653786
hsa-miR-17-3p27663787
hsa-miR-17-5p27673788
hsa-miR-181a-2-3p27683789
hsa-miR-181a-3p27693790
hsa-miR-181a-5p27703791
hsa-miR-181b-3p27713792
hsa-miR-181b-5p27723793
hsa-miR-181c-3p27733794
hsa-miR-181c-5p27743795
hsa-miR-181d27753796
hsa-miR-182-3p27763797
hsa-miR-182527773798
hsa-miR-182-5p27783799
hsa-miR-182727793800
hsa-miR-183-3p27803801
hsa-miR-183-5p27813802
hsa-miR-18427823803
hsa-miR-185-3p27833804
hsa-miR-185-5p27843805
hsa-miR-186-3p27853806
hsa-miR-186-5p27863807
hsa-miR-187-3p27873808
hsa-miR-187-5p27883809
hsa-miR-188-3p27893810
hsa-miR-188-5p27903811
hsa-miR-18a-3p27913812
hsa-miR-18a-5p27923813
hsa-miR-18b-3p27933814
hsa-miR-18b-5p27943815
hsa-miR-190827953816
hsa-miR-1909-3p27963817
hsa-miR-1909-5p27973818
hsa-miR-190a27983819
hsa-miR-190b27993820
hsa-miR-191028003821
hsa-miR-1911-3p28013822
hsa-miR-1911-5p28023823
hsa-miR-191228033824
hsa-miR-191328043825
hsa-miR-191-3p28053826
hsa-miR-1914-3p28063827
hsa-miR-1914-5p28073828
hsa-miR-1915-3p28083829
hsa-miR-1915-5p28093830
hsa-miR-191-5p28103831
hsa-miR-192-3p28113832
hsa-miR-192-5p28123833
hsa-miR-193a-3p28133834
hsa-miR-193a-5p28143835
hsa-miR-193b-3p28153836
hsa-miR-193b-5p28163837
hsa-miR-194-3p28173838
hsa-miR-194-5p28183839
hsa-miR-195-3p28193840
hsa-miR-195-5p28203841
hsa-miR-196a-3p28213842
hsa-miR-196a-5p28223843
hsa-miR-196b-3p28233844
hsa-miR-196b-5p28243845
hsa-miR-197228253846
hsa-miR-197328263847
hsa-miR-197-3p28273848
hsa-miR-197-5p28283849
hsa-miR-197628293850
hsa-miR-19828303851
hsa-miR-199a-3p28313852
hsa-miR-199a-5p28323853
hsa-miR-199b-3p28333854
hsa-miR-199b-5p28343855
hsa-miR-19a-3p28353856
hsa-miR-19a-5p28363857
hsa-miR-19b-1-5p28373858
hsa-miR-19b-2-5p28383859
hsa-miR-19b-3p28393860
hsa-miR-200a-3p28403861
hsa-miR-200a-5p28413862
hsa-miR-200b-3p28423863
hsa-miR-200b-5p28433864
hsa-miR-200c-3p28443865
hsa-miR-200c-5p28453866
hsa-miR-202-3p28463867
hsa-miR-202-5p28473868
hsa-miR-203a28483869
hsa-miR-203b-3p28493870
hsa-miR-203b-5p28503871
hsa-miR-204-3p28513872
hsa-miR-204-5p28523873
hsa-miR-205228533874
hsa-miR-205328543875
hsa-miR-205-3p28553876
hsa-miR-205428563877
hsa-miR-205-5p28573878
hsa-miR-20628583879
hsa-miR-208a28593880
hsa-miR-208b28603881
hsa-miR-20a-3p28613882
hsa-miR-20a-5p28623883
hsa-miR-20b-3p28633884
hsa-miR-20b-5p28643885
hsa-miR-21028653886
hsa-miR-211028663887
hsa-miR-211328673888
hsa-miR-211-3p28683889
hsa-miR-2114-3p28693890
hsa-miR-2114-5p28703891
hsa-miR-2115-3p28713892
hsa-miR-2115-5p28723893
hsa-miR-211-5p28733894
hsa-miR-2116-3p28743895
hsa-miR-2116-5p28753896
hsa-miR-211728763897
hsa-miR-212-3p28773898
hsa-miR-212-5p28783899
hsa-miR-21-3p28793900
hsa-miR-214-3p28803901
hsa-miR-214-5p28813902
hsa-miR-21528823903
hsa-miR-21-5p28833904
hsa-miR-216a-3p28843905
hsa-miR-216a-5p28853906
hsa-miR-216b28863907
hsa-miR-21728873908
hsa-miR-218-1-3p28883909
hsa-miR-218-2-3p28893910
hsa-miR-218-5p28903911
hsa-miR-219-1-3p28913912
hsa-miR-219-2-3p28923913
hsa-miR-219-5p28933914
hsa-miR-221-3p28943915
hsa-miR-221-5p28953916
hsa-miR-222-3p28963917
hsa-miR-222-5p28973918
hsa-miR-223-3p28983919
hsa-miR-223-5p28993920
hsa-miR-22-3p29003921
hsa-miR-224-3p29013922
hsa-miR-224-5p29023923
hsa-miR-22-5p29033924
hsa-miR-227629043925
hsa-miR-2277-3p29053926
hsa-miR-2277-5p29063927
hsa-miR-227829073928
hsa-miR-2355-3p29083929
hsa-miR-2355-5p29093930
hsa-miR-239229103931
hsa-miR-23a-3p29113932
hsa-miR-23a-5p29123933
hsa-miR-23b-3p29133934
hsa-miR-23b-5p29143935
hsa-miR-23c29153936
hsa-miR-24-1-5p29163937
hsa-miR-24-2-5p29173938
hsa-miR-24-3p29183939
hsa-miR-2467-3p29193940
hsa-miR-2467-5p29203941
hsa-miR-25-3p29213942
hsa-miR-25-5p29223943
hsa-miR-2681-3p29233944
hsa-miR-2681-5p29243945
hsa-miR-2682-3p29253946
hsa-miR-2682-5p29263947
hsa-miR-26a-1-3p29273948
hsa-miR-26a-2-3p29283949
hsa-miR-26a-5p29293950
hsa-miR-26b-3p29303951
hsa-miR-26b-5p29313952
hsa-miR-27a-3p29323953
hsa-miR-27a-5p29333954
hsa-miR-27b-3p29343955
hsa-miR-27b-5p29353956
hsa-miR-28-3p29363957
hsa-miR-28-5p29373958
hsa-miR-286129383959
hsa-miR-290929393960
hsa-miR-296-3p29403961
hsa-miR-2964a-3p29413962
hsa-miR-2964a-5p29423963
hsa-miR-296-5p29433964
hsa-miR-29729443965
hsa-miR-29829453966
hsa-miR-299-3p29463967
hsa-miR-299-5p29473968
hsa-miR-29a-3p29483969
hsa-miR-29a-5p29493970
hsa-miR-29b-1-5p29503971
hsa-miR-29b-2-5p29513972
hsa-miR-29b-3p29523973
hsa-miR-29c-3p29533974
hsa-miR-29c-5p29543975
hsa-miR-30029553976
hsa-miR-301a-3p29563977
hsa-miR-301a-5p29573978
hsa-miR-301b29583979
hsa-miR-302a-3p29593980
hsa-miR-302a-5p29603981
hsa-miR-302b-3p29613982
hsa-miR-302b-5p29623983
hsa-miR-302c-3p29633984
hsa-miR-302c-5p29643985
hsa-miR-302d-3p29653986
hsa-miR-302d-5p29663987
hsa-miR-302e29673988
hsa-miR-302f29683989
hsa-miR-3064-3p29693990
hsa-miR-3064-5p29703991
hsa-miR-3065-3p29713992
hsa-miR-3065-5p29723993
hsa-miR-3074-3p29733994
hsa-miR-3074-5p29743995
hsa-miR-30a-3p29753996
hsa-miR-30a-5p29763997
hsa-miR-30b-3p29773998
hsa-miR-30b-5p29783999
hsa-miR-30c-1-3p29794000
hsa-miR-30c-2-3p29804001
hsa-miR-30c-5p29814002
hsa-miR-30d-3p29824003
hsa-miR-30d-5p29834004
hsa-miR-30e-3p29844005
hsa-miR-30e-5p29854006
hsa-miR-311529864007
hsa-miR-311629874008
hsa-miR-3117-3p29884009
hsa-miR-3117-5p29894010
hsa-miR-311829904011
hsa-miR-311929914012
hsa-miR-3120-3p29924013
hsa-miR-3120-5p29934014
hsa-miR-3121-3p29944015
hsa-miR-3121-5p29954016
hsa-miR-312229964017
hsa-miR-312329974018
hsa-miR-3124-3p29984019
hsa-miR-3124-5p29994020
hsa-miR-312530004021
hsa-miR-3126-3p30014022
hsa-miR-3126-5p30024023
hsa-miR-3127-3p30034024
hsa-miR-3127-5p30044025
hsa-miR-312830054026
hsa-miR-3129-3p30064027
hsa-miR-3129-5p30074028
hsa-miR-3130-3p30084029
hsa-miR-3130-5p30094030
hsa-miR-313130104031
hsa-miR-313230114032
hsa-miR-313330124033
hsa-miR-313430134034
hsa-miR-3135a30144035
hsa-miR-3135b30154036
hsa-miR-3136-3p30164037
hsa-miR-3136-5p30174038
hsa-miR-313730184039
hsa-miR-313830194040
hsa-miR-313930204041
hsa-miR-31-3p30214042
hsa-miR-3140-3p30224043
hsa-miR-3140-5p30234044
hsa-miR-314130244045
hsa-miR-314230254046
hsa-miR-314330264047
hsa-miR-3144-3p30274048
hsa-miR-3144-5p30284049
hsa-miR-3145-3p30294050
hsa-miR-3145-5p30304051
hsa-miR-314630314052
hsa-miR-314730324053
hsa-miR-314830334054
hsa-miR-314930344055
hsa-miR-3150a-3p30354056
hsa-miR-3150a-5p30364057
hsa-miR-3150b-3p30374058
hsa-miR-3150b-5p30384059
hsa-miR-315130394060
hsa-miR-3152-3p30404061
hsa-miR-3152-5p30414062
hsa-miR-315330424063
hsa-miR-315430434064
hsa-miR-3155a30444065
hsa-miR-3155b30454066
hsa-miR-3156-3p30464067
hsa-miR-3156-5p30474068
hsa-miR-3157-3p30484069
hsa-miR-3157-5p30494070
hsa-miR-3158-3p30504071
hsa-miR-3158-5p30514072
hsa-miR-315930524073
hsa-miR-31-5p30534074
hsa-miR-3160-3p30544075
hsa-miR-3160-5p30554076
hsa-miR-316130564077
hsa-miR-3162-3p30574078
hsa-miR-3162-5p30584079
hsa-miR-316330594080
hsa-miR-316430604081
hsa-miR-316530614082
hsa-miR-316630624083
hsa-miR-316730634084
hsa-miR-316830644085
hsa-miR-316930654086
hsa-miR-317030664087
hsa-miR-317130674088
hsa-miR-3173-3p30684089
hsa-miR-3173-5p30694090
hsa-miR-317430704091
hsa-miR-317530714092
hsa-miR-317630724093
hsa-miR-3177-3p30734094
hsa-miR-3177-5p30744095
hsa-miR-317830754096
hsa-miR-317930764097
hsa-miR-318030774098
hsa-miR-3180-3p30784099
hsa-miR-3180-5p30794100
hsa-miR-318130804101
hsa-miR-318230814102
hsa-miR-318330824103
hsa-miR-3184-3p30834104
hsa-miR-3184-5p30844105
hsa-miR-318530854106
hsa-miR-3186-3p30864107
hsa-miR-3186-5p30874108
hsa-miR-3187-3p30884109
hsa-miR-3187-5p30894110
hsa-miR-318830904111
hsa-miR-3189-3p30914112
hsa-miR-3189-5p30924113
hsa-miR-3190-3p30934114
hsa-miR-3190-5p30944115
hsa-miR-3191-3p30954116
hsa-miR-3191-5p30964117
hsa-miR-319230974118
hsa-miR-319330984119
hsa-miR-3194-3p30994120
hsa-miR-3194-5p31004121
hsa-miR-319531014122
hsa-miR-319631024123
hsa-miR-319731034124
hsa-miR-319831044125
hsa-miR-319931054126
hsa-miR-3200-3p31064127
hsa-miR-3200-5p31074128
hsa-miR-320131084129
hsa-miR-320231094130
hsa-miR-320a31104131
hsa-miR-320b31114132
hsa-miR-320c31124133
hsa-miR-320d31134134
hsa-miR-320e31144135
hsa-miR-323a-3p31154136
hsa-miR-323a-5p31164137
hsa-miR-323b-3p31174138
hsa-miR-323b-5p31184139
hsa-miR-32-3p31194140
hsa-miR-324-3p31204141
hsa-miR-324-5p31214142
hsa-miR-32531224143
hsa-miR-32-5p31234144
hsa-miR-32631244145
hsa-miR-32831254146
hsa-miR-32931264147
hsa-miR-330-3p31274148
hsa-miR-330-5p31284149
hsa-miR-331-3p31294150
hsa-miR-331-5p31304151
hsa-miR-335-3p31314152
hsa-miR-335-5p31324153
hsa-miR-337-3p31334154
hsa-miR-337-5p31344155
hsa-miR-338-3p31354156
hsa-miR-338-5p31364157
hsa-miR-339-3p31374158
hsa-miR-339-5p31384159
hsa-miR-33a-3p31394160
hsa-miR-33a-5p31404161
hsa-miR-33b-3p31414162
hsa-miR-33b-5p31424163
hsa-miR-340-3p31434164
hsa-miR-340-5p31444165
hsa-miR-342-3p31454166
hsa-miR-342-5p31464167
hsa-miR-345-3p31474168
hsa-miR-345-5p31484169
hsa-miR-34631494170
hsa-miR-34a-3p31504171
hsa-miR-34a-5p31514172
hsa-miR-34b-3p31524173
hsa-miR-34b-5p31534174
hsa-miR-34c-3p31544175
hsa-miR-34c-5p31554176
hsa-miR-3529-3p31564177
hsa-miR-3529-5p31574178
hsa-miR-3591-3p31584179
hsa-miR-3591-5p31594180
hsa-miR-3605-3p31604181
hsa-miR-3605-5p31614182
hsa-miR-3606-3p31624183
hsa-miR-3606-5p31634184
hsa-miR-3607-3p31644185
hsa-miR-3607-5p31654186
hsa-miR-360931664187
hsa-miR-361031674188
hsa-miR-361131684189
hsa-miR-361231694190
hsa-miR-3613-3p31704191
hsa-miR-3613-5p31714192
hsa-miR-361-3p31724193
hsa-miR-3614-3p31734194
hsa-miR-3614-5p31744195
hsa-miR-361531754196
hsa-miR-361-5p31764197
hsa-miR-3616-3p31774198
hsa-miR-3616-5p31784199
hsa-miR-3617-3p31794200
hsa-miR-3617-5p31804201
hsa-miR-361831814202
hsa-miR-3619-3p31824203
hsa-miR-3619-5p31834204
hsa-miR-3620-3p31844205
hsa-miR-3620-5p31854206
hsa-miR-362131864207
hsa-miR-3622a-3p31874208
hsa-miR-3622a-5p31884209
hsa-miR-3622b-3p31894210
hsa-miR-3622b-5p31904211
hsa-miR-362-3p31914212
hsa-miR-362-5p31924213
hsa-miR-363-3p31934214
hsa-miR-363-5p31944215
hsa-miR-364631954216
hsa-miR-364831964217
hsa-miR-364931974218
hsa-miR-365031984219
hsa-miR-365131994220
hsa-miR-365232004221
hsa-miR-365332014222
hsa-miR-365432024223
hsa-miR-365532034224
hsa-miR-365632044225
hsa-miR-365732054226
hsa-miR-365832064227
hsa-miR-365932074228
hsa-miR-365a-3p32084229
hsa-miR-365a-5p32094230
hsa-miR-365b-3p32104231
hsa-miR-365b-5p32114232
hsa-miR-366032124233
hsa-miR-366132134234
hsa-miR-366232144235
hsa-miR-3663-3p32154236
hsa-miR-3663-5p32164237
hsa-miR-3664-3p32174238
hsa-miR-3664-5p32184239
hsa-miR-366532194240
hsa-miR-366632204241
hsa-miR-3667-3p32214242
hsa-miR-3667-5p32224243
hsa-miR-366832234244
hsa-miR-366932244245
hsa-miR-367032254246
hsa-miR-367132264247
hsa-miR-367232274248
hsa-miR-367332284249
hsa-miR-367-3p32294250
hsa-miR-367432304251
hsa-miR-3675-3p32314252
hsa-miR-3675-5p32324253
hsa-miR-367-5p32334254
hsa-miR-3676-3p32344255
hsa-miR-3676-5p32354256
hsa-miR-3677-3p32364257
hsa-miR-3677-5p32374258
hsa-miR-3678-3p32384259
hsa-miR-3678-5p32394260
hsa-miR-3679-3p32404261
hsa-miR-3679-5p32414262
hsa-miR-3680-3p32424263
hsa-miR-3680-5p32434264
hsa-miR-3681-3p32444265
hsa-miR-3681-5p32454266
hsa-miR-3682-3p32464267
hsa-miR-3682-5p32474268
hsa-miR-368332484269
hsa-miR-368432494270
hsa-miR-368532504271
hsa-miR-368632514272
hsa-miR-368732524273
hsa-miR-3688-3p32534274
hsa-miR-3688-5p32544275
hsa-miR-3689a-3p32554276
hsa-miR-3689a-5p32564277
hsa-miR-3689b-3p32574278
hsa-miR-3689b-5p32584279
hsa-miR-3689c32594280
hsa-miR-3689d32604281
hsa-miR-3689e32614282
hsa-miR-3689f32624283
hsa-miR-369032634284
hsa-miR-3691-3p32644285
hsa-miR-3691-5p32654286
hsa-miR-3692-3p32664287
hsa-miR-3692-5p32674288
hsa-miR-369-3p32684289
hsa-miR-369-5p32694290
hsa-miR-37032704291
hsa-miR-371332714292
hsa-miR-371432724293
hsa-miR-371a-3p32734294
hsa-miR-371a-5p32744295
hsa-miR-371b-3p32754296
hsa-miR-371b-5p32764297
hsa-miR-37232774298
hsa-miR-373-3p32784299
hsa-miR-373-5p32794300
hsa-miR-374a-3p32804301
hsa-miR-374a-5p32814302
hsa-miR-374b-3p32824303
hsa-miR-374b-5p32834304
hsa-miR-374c-3p32844305
hsa-miR-374c-5p32854306
hsa-miR-37532864307
hsa-miR-376a-2-5p32874308
hsa-miR-376a-3p32884309
hsa-miR-376a-5p32894310
hsa-miR-376b-3p32904311
hsa-miR-376b-5p32914312
hsa-miR-376c-3p32924313
hsa-miR-376c-5p32934314
hsa-miR-377-3p32944315
hsa-miR-377-5p32954316
hsa-miR-378a-3p32964317
hsa-miR-378a-5p32974318
hsa-miR-378b32984319
hsa-miR-378c32994320
hsa-miR-378d33004321
hsa-miR-378e33014322
hsa-miR-378f33024323
hsa-miR-378g33034324
hsa-miR-378h33044325
hsa-miR-378i33054326
hsa-miR-378j33064327
hsa-miR-379-3p33074328
hsa-miR-379-5p33084329
hsa-miR-380-3p33094330
hsa-miR-380-5p33104331
hsa-miR-381-3p33114332
hsa-miR-381-5p33124333
hsa-miR-382-3p33134334
hsa-miR-382-5p33144335
hsa-miR-38333154336
hsa-miR-38433164337
hsa-miR-390733174338
hsa-miR-390833184339
hsa-miR-390933194340
hsa-miR-391033204341
hsa-miR-391133214342
hsa-miR-391233224343
hsa-miR-3913-3p33234344
hsa-miR-3913-5p33244345
hsa-miR-391433254346
hsa-miR-391533264347
hsa-miR-391633274348
hsa-miR-391733284349
hsa-miR-391833294350
hsa-miR-391933304351
hsa-miR-392033314352
hsa-miR-392133324353
hsa-miR-3922-3p33334354
hsa-miR-3922-5p33344355
hsa-miR-392333354356
hsa-miR-392433364357
hsa-miR-3925-3p33374358
hsa-miR-3925-5p33384359
hsa-miR-392633394360
hsa-miR-3927-3p33404361
hsa-miR-3927-5p33414362
hsa-miR-392833424363
hsa-miR-392933434364
hsa-miR-3934-3p33444365
hsa-miR-3934-5p33454366
hsa-miR-393533464367
hsa-miR-393633474368
hsa-miR-393733484369
hsa-miR-393833494370
hsa-miR-393933504371
hsa-miR-3940-3p33514372
hsa-miR-3940-5p33524373
hsa-miR-394133534374
hsa-miR-3942-3p33544375
hsa-miR-3942-5p33554376
hsa-miR-394333564377
hsa-miR-3944-3p33574378
hsa-miR-3944-5p33584379
hsa-miR-394533594380
hsa-miR-396033604381
hsa-miR-397233614382
hsa-miR-397333624383
hsa-miR-397433634384
hsa-miR-397533644385
hsa-miR-397633654386
hsa-miR-397733664387
hsa-miR-397833674388
hsa-miR-409-3p33684389
hsa-miR-409-5p33694390
hsa-miR-41033704391
hsa-miR-411-3p33714392
hsa-miR-411-5p33724393
hsa-miR-41233734394
hsa-miR-42133744395
hsa-miR-422a33754396
hsa-miR-423-3p33764397
hsa-miR-423-5p33774398
hsa-miR-424-3p33784399
hsa-miR-424-5p33794400
hsa-miR-425133804401
hsa-miR-425233814402
hsa-miR-425333824403
hsa-miR-425-3p33834404
hsa-miR-425433844405
hsa-miR-425533854406
hsa-miR-425-5p33864407
hsa-miR-425633874408
hsa-miR-425733884409
hsa-miR-425833894410
hsa-miR-425933904411
hsa-miR-426033914412
hsa-miR-426133924413
hsa-miR-426233934414
hsa-miR-426333944415
hsa-miR-426433954416
hsa-miR-426533964417
hsa-miR-426633974418
hsa-miR-426733984419
hsa-miR-426833994420
hsa-miR-426934004421
hsa-miR-427034014422
hsa-miR-427134024423
hsa-miR-427234034424
hsa-miR-427334044425
hsa-miR-427434054426
hsa-miR-427534064427
hsa-miR-427634074428
hsa-miR-427734084429
hsa-miR-427834094430
hsa-miR-427934104431
hsa-miR-428034114432
hsa-miR-428134124433
hsa-miR-428234134434
hsa-miR-428334144435
hsa-miR-428434154436
hsa-miR-428534164437
hsa-miR-428634174438
hsa-miR-428734184439
hsa-miR-428834194440
hsa-miR-428934204441
hsa-miR-42934214442
hsa-miR-429034224443
hsa-miR-429134234444
hsa-miR-429234244445
hsa-miR-429334254446
hsa-miR-429434264447
hsa-miR-429534274448
hsa-miR-429634284449
hsa-miR-429734294450
hsa-miR-429834304451
hsa-miR-429934314452
hsa-miR-430034324453
hsa-miR-430134334454
hsa-miR-430234344455
hsa-miR-430334354456
hsa-miR-430434364457
hsa-miR-430534374458
hsa-miR-430634384459
hsa-miR-430734394460
hsa-miR-430834404461
hsa-miR-430934414462
hsa-miR-431034424463
hsa-miR-431134434464
hsa-miR-431234444465
hsa-miR-431334454466
hsa-miR-431-3p34464467
hsa-miR-431434474468
hsa-miR-431534484469
hsa-miR-431-5p34494470
hsa-miR-431634504471
hsa-miR-431734514472
hsa-miR-431834524473
hsa-miR-431934534474
hsa-miR-432034544475
hsa-miR-432134554476
hsa-miR-432234564477
hsa-miR-432334574478
hsa-miR-432-3p34584479
hsa-miR-432434594480
hsa-miR-432534604481
hsa-miR-432-5p34614482
hsa-miR-432634624483
hsa-miR-432734634484
hsa-miR-432834644485
hsa-miR-432934654486
hsa-miR-43334664487
hsa-miR-433034674488
hsa-miR-441734684489
hsa-miR-441834694490
hsa-miR-4419a34704491
hsa-miR-4419b34714492
hsa-miR-442034724493
hsa-miR-442134734494
hsa-miR-442234744495
hsa-miR-4423-3p34754496
hsa-miR-4423-5p34764497
hsa-miR-442434774498
hsa-miR-442534784499
hsa-miR-442634794500
hsa-miR-442734804501
hsa-miR-442834814502
hsa-miR-442934824503
hsa-miR-443034834504
hsa-miR-443134844505
hsa-miR-443234854506
hsa-miR-4433-3p34864507
hsa-miR-4433-5p34874508
hsa-miR-443434884509
hsa-miR-443534894510
hsa-miR-4436a34904511
hsa-miR-4436b-3p34914512
hsa-miR-4436b-5p34924513
hsa-miR-443734934514
hsa-miR-443834944515
hsa-miR-443934954516
hsa-miR-444034964517
hsa-miR-444134974518
hsa-miR-444234984519
hsa-miR-444334994520
hsa-miR-444435004521
hsa-miR-4445-3p35014522
hsa-miR-4445-5p35024523
hsa-miR-4446-3p35034524
hsa-miR-4446-5p35044525
hsa-miR-444735054526
hsa-miR-444835064527
hsa-miR-444935074528
hsa-miR-445035084529
hsa-miR-445135094530
hsa-miR-445235104531
hsa-miR-445335114532
hsa-miR-445435124533
hsa-miR-445535134534
hsa-miR-445635144535
hsa-miR-445735154536
hsa-miR-445835164537
hsa-miR-445935174538
hsa-miR-446035184539
hsa-miR-446135194540
hsa-miR-446235204541
hsa-miR-446335214542
hsa-miR-446435224543
hsa-miR-446535234544
hsa-miR-446635244545
hsa-miR-446735254546
hsa-miR-446835264547
hsa-miR-446935274548
hsa-miR-447035284549
hsa-miR-447145505571
hsa-miR-447245515572
hsa-miR-447345525573
hsa-miR-4474-3p45535574
hsa-miR-4474-5p45545575
hsa-miR-447545555576
hsa-miR-447645565577
hsa-miR-4477a45575578
hsa-miR-4477b45585579
hsa-miR-447845595580
hsa-miR-447945605581
hsa-miR-44845615582
hsa-miR-448045625583
hsa-miR-448145635584
hsa-miR-4482-3p45645585
hsa-miR-4482-5p45655586
hsa-miR-448345665587
hsa-miR-448445675588
hsa-miR-448545685589
hsa-miR-448645695590
hsa-miR-448745705591
hsa-miR-448845715592
hsa-miR-448945725593
hsa-miR-449045735594
hsa-miR-449145745595
hsa-miR-449245755596
hsa-miR-449345765597
hsa-miR-449445775598
hsa-miR-449545785599
hsa-miR-449645795600
hsa-miR-449745805601
hsa-miR-449845815602
hsa-miR-449945825603
hsa-miR-449a45835604
hsa-miR-449b-3p45845605
hsa-miR-449b-5p45855606
hsa-miR-449c-3p45865607
hsa-miR-449c-5p45875608
hsa-miR-450045885609
hsa-miR-450145895610
hsa-miR-450245905611
hsa-miR-450345915612
hsa-miR-450445925613
hsa-miR-450545935614
hsa-miR-450645945615
hsa-miR-450745955616
hsa-miR-450845965617
hsa-miR-450945975618
hsa-miR-450a-3p45985619
hsa-miR-450a-5p45995620
hsa-miR-450b-3p46005621
hsa-miR-450b-5p46015622
hsa-miR-451046025623
hsa-miR-451146035624
hsa-miR-451246045625
hsa-miR-451346055626
hsa-miR-451446065627
hsa-miR-451546075628
hsa-miR-451646085629
hsa-miR-451746095630
hsa-miR-451846105631
hsa-miR-451946115632
hsa-miR-451a46125633
hsa-miR-451b46135634
hsa-miR-4520a-3p46145635
hsa-miR-4520a-5p46155636
hsa-miR-4520b-3p46165637
hsa-miR-4520b-5p46175638
hsa-miR-452146185639
hsa-miR-452246195640
hsa-miR-452346205641
hsa-miR-452-3p46215642
hsa-miR-4524a-3p46225643
hsa-miR-4524a-5p46235644
hsa-miR-4524b-3p46245645
hsa-miR-4524b-5p46255646
hsa-miR-452546265647
hsa-miR-452-5p46275648
hsa-miR-452646285649
hsa-miR-452746295650
hsa-miR-452846305651
hsa-miR-4529-3p46315652
hsa-miR-4529-5p46325653
hsa-miR-453046335654
hsa-miR-453146345655
hsa-miR-453246355656
hsa-miR-453346365657
hsa-miR-453446375658
hsa-miR-453546385659
hsa-miR-4536-3p46395660
hsa-miR-4536-5p46405661
hsa-miR-453746415662
hsa-miR-453846425663
hsa-miR-453946435664
hsa-miR-454046445665
hsa-miR-454-3p46455666
hsa-miR-454-5p46465667
hsa-miR-455-3p46475668
hsa-miR-455-5p46485669
hsa-miR-4632-3p46495670
hsa-miR-4632-5p46505671
hsa-miR-4633-3p46515672
hsa-miR-4633-5p46525673
hsa-miR-463446535674
hsa-miR-463546545675
hsa-miR-463646555676
hsa-miR-463746565677
hsa-miR-4638-3p46575678
hsa-miR-4638-5p46585679
hsa-miR-4639-3p46595680
hsa-miR-4639-5p46605681
hsa-miR-4640-3p46615682
hsa-miR-4640-5p46625683
hsa-miR-464146635684
hsa-miR-464246645685
hsa-miR-464346655686
hsa-miR-464446665687
hsa-miR-4645-3p46675688
hsa-miR-4645-5p46685689
hsa-miR-4646-3p46695690
hsa-miR-4646-5p46705691
hsa-miR-464746715692
hsa-miR-464846725693
hsa-miR-4649-3p46735694
hsa-miR-4649-5p46745695
hsa-miR-4650-3p46755696
hsa-miR-4650-5p46765697
hsa-miR-465146775698
hsa-miR-4652-3p46785699
hsa-miR-4652-5p46795700
hsa-miR-4653-3p46805701
hsa-miR-4653-5p46815702
hsa-miR-465446825703
hsa-miR-4655-3p46835704
hsa-miR-4655-5p46845705
hsa-miR-465646855706
hsa-miR-465746865707
hsa-miR-465846875708
hsa-miR-4659a-3p46885709
hsa-miR-4659a-5p46895710
hsa-miR-4659b-3p46905711
hsa-miR-4659b-5p46915712
hsa-miR-46646925713
hsa-miR-466046935714
hsa-miR-4661-3p46945715
hsa-miR-4661-5p46955716
hsa-miR-4662a-3p46965717
hsa-miR-4662a-5p46975718
hsa-miR-4662b46985719
hsa-miR-466346995720
hsa-miR-4664-3p47005721
hsa-miR-4664-5p47015722
hsa-miR-4665-3p47025723
hsa-miR-4665-5p47035724
hsa-miR-4666a-3p47045725
hsa-miR-4666a-5p47055726
hsa-miR-4666b47065727
hsa-miR-4667-3p47075728
hsa-miR-4667-5p47085729
hsa-miR-4668-3p47095730
hsa-miR-4668-5p47105731
hsa-miR-466947115732
hsa-miR-4670-3p47125733
hsa-miR-4670-5p47135734
hsa-miR-4671-3p47145735
hsa-miR-4671-5p47155736
hsa-miR-467247165737
hsa-miR-467347175738
hsa-miR-467447185739
hsa-miR-467547195740
hsa-miR-4676-3p47205741
hsa-miR-4676-5p47215742
hsa-miR-4677-3p47225743
hsa-miR-4677-5p47235744
hsa-miR-467847245745
hsa-miR-467947255746
hsa-miR-4680-3p47265747
hsa-miR-4680-5p47275748
hsa-miR-468147285749
hsa-miR-468247295750
hsa-miR-468347305751
hsa-miR-4684-3p47315752
hsa-miR-4684-5p47325753
hsa-miR-4685-3p47335754
hsa-miR-4685-5p47345755
hsa-miR-468647355756
hsa-miR-4687-3p47365757
hsa-miR-4687-5p47375758
hsa-miR-468847385759
hsa-miR-468947395760
hsa-miR-4690-3p47405761
hsa-miR-4690-5p47415762
hsa-miR-4691-3p47425763
hsa-miR-4691-5p47435764
hsa-miR-469247445765
hsa-miR-4693-3p47455766
hsa-miR-4693-5p47465767
hsa-miR-4694-3p47475768
hsa-miR-4694-5p47485769
hsa-miR-4695-3p47495770
hsa-miR-4695-5p47505771
hsa-miR-469647515772
hsa-miR-4697-3p47525773
hsa-miR-4697-5p47535774
hsa-miR-469847545775
hsa-miR-4699-3p47555776
hsa-miR-4699-5p47565777
hsa-miR-4700-3p47575778
hsa-miR-4700-5p47585779
hsa-miR-4701-3p47595780
hsa-miR-4701-5p47605781
hsa-miR-4703-3p47615782
hsa-miR-4703-5p47625783
hsa-miR-4704-3p47635784
hsa-miR-4704-5p47645785
hsa-miR-470547655786
hsa-miR-470647665787
hsa-miR-4707-3p47675788
hsa-miR-4707-5p47685789
hsa-miR-4708-3p47695790
hsa-miR-4708-5p47705791
hsa-miR-4709-3p47715792
hsa-miR-4709-5p47725793
hsa-miR-471047735794
hsa-miR-4711-3p47745795
hsa-miR-4711-5p47755796
hsa-miR-4712-3p47765797
hsa-miR-4712-5p47775798
hsa-miR-4713-3p47785799
hsa-miR-4713-5p47795800
hsa-miR-4714-3p47805801
hsa-miR-4714-5p47815802
hsa-miR-4715-3p47825803
hsa-miR-4715-5p47835804
hsa-miR-4716-3p47845805
hsa-miR-4716-5p47855806
hsa-miR-4717-3p47865807
hsa-miR-4717-5p47875808
hsa-miR-471847885809
hsa-miR-471947895810
hsa-miR-4720-3p47905811
hsa-miR-4720-5p47915812
hsa-miR-472147925813
hsa-miR-4722-3p47935814
hsa-miR-4722-5p47945815
hsa-miR-4723-3p47955816
hsa-miR-4723-5p47965817
hsa-miR-4724-3p47975818
hsa-miR-4724-5p47985819
hsa-miR-4725-3p47995820
hsa-miR-4725-5p48005821
hsa-miR-4726-3p48015822
hsa-miR-4726-5p48025823
hsa-miR-4727-3p48035824
hsa-miR-4727-5p48045825
hsa-miR-4728-3p48055826
hsa-miR-4728-5p48065827
hsa-miR-472948075828
hsa-miR-473048085829
hsa-miR-4731-3p48095830
hsa-miR-4731-5p48105831
hsa-miR-4732-3p48115832
hsa-miR-4732-5p48125833
hsa-miR-4733-3p48135834
hsa-miR-4733-5p48145835
hsa-miR-473448155836
hsa-miR-4735-3p48165837
hsa-miR-4735-5p48175838
hsa-miR-473648185839
hsa-miR-473748195840
hsa-miR-4738-3p48205841
hsa-miR-4738-5p48215842
hsa-miR-473948225843
hsa-miR-4740-3p48235844
hsa-miR-4740-5p48245845
hsa-miR-474148255846
hsa-miR-4742-3p48265847
hsa-miR-4742-5p48275848
hsa-miR-4743-3p48285849
hsa-miR-4743-5p48295850
hsa-miR-474448305851
hsa-miR-4745-3p48315852
hsa-miR-4745-5p48325853
hsa-miR-4746-3p48335854
hsa-miR-4746-5p48345855
hsa-miR-4747-3p48355856
hsa-miR-4747-5p48365857
hsa-miR-474848375858
hsa-miR-4749-3p48385859
hsa-miR-4749-5p48395860
hsa-miR-4750-3p48405861
hsa-miR-4750-5p48415862
hsa-miR-475148425863
hsa-miR-475248435864
hsa-miR-4753-3p48445865
hsa-miR-4753-5p48455866
hsa-miR-475448465867
hsa-miR-4755-3p48475868
hsa-miR-4755-5p48485869
hsa-miR-4756-3p48495870
hsa-miR-4756-5p48505871
hsa-miR-4757-3p48515872
hsa-miR-4757-5p48525873
hsa-miR-4758-3p48535874
hsa-miR-4758-5p48545875
hsa-miR-475948555876
hsa-miR-4760-3p48565877
hsa-miR-4760-5p48575878
hsa-miR-4761-3p48585879
hsa-miR-4761-5p48595880
hsa-miR-4762-3p48605881
hsa-miR-4762-5p48615882
hsa-miR-4763-3p48625883
hsa-miR-4763-5p48635884
hsa-miR-4764-3p48645885
hsa-miR-4764-5p48655886
hsa-miR-476548665887
hsa-miR-4766-3p48675888
hsa-miR-4766-5p48685889
hsa-miR-476748695890
hsa-miR-4768-3p48705891
hsa-miR-4768-5p48715892
hsa-miR-4769-3p48725893
hsa-miR-4769-5p48735894
hsa-miR-477048745895
hsa-miR-477148755896
hsa-miR-4772-3p48765897
hsa-miR-4772-5p48775898
hsa-miR-477348785899
hsa-miR-4774-3p48795900
hsa-miR-4774-5p48805901
hsa-miR-477548815902
hsa-miR-4776-3p48825903
hsa-miR-4776-5p48835904
hsa-miR-4777-3p48845905
hsa-miR-4777-5p48855906
hsa-miR-4778-3p48865907
hsa-miR-4778-5p48875908
hsa-miR-477948885909
hsa-miR-478048895910
hsa-miR-4781-3p48905911
hsa-miR-4781-5p48915912
hsa-miR-4782-3p48925913
hsa-miR-4782-5p48935914
hsa-miR-4783-3p48945915
hsa-miR-4783-5p48955916
hsa-miR-478448965917
hsa-miR-478548975918
hsa-miR-4786-3p48985919
hsa-miR-4786-5p48995920
hsa-miR-4787-3p49005921
hsa-miR-4787-5p49015922
hsa-miR-478849025923
hsa-miR-4789-3p49035924
hsa-miR-4789-5p49045925
hsa-miR-4790-3p49055926
hsa-miR-4790-5p49065927
hsa-miR-479149075928
hsa-miR-479249085929
hsa-miR-4793-3p49095930
hsa-miR-4793-5p49105931
hsa-miR-479449115932
hsa-miR-4795-3p49125933
hsa-miR-4795-5p49135934
hsa-miR-4796-3p49145935
hsa-miR-4796-5p49155936
hsa-miR-4797-3p49165937
hsa-miR-4797-5p49175938
hsa-miR-4798-3p49185939
hsa-miR-4798-5p49195940
hsa-miR-4799-3p49205941
hsa-miR-4799-5p49215942
hsa-miR-4800-3p49225943
hsa-miR-4800-5p49235944
hsa-miR-480149245945
hsa-miR-4802-3p49255946
hsa-miR-4802-5p49265947
hsa-miR-480349275948
hsa-miR-4804-3p49285949
hsa-miR-4804-5p49295950
hsa-miR-483-3p49305951
hsa-miR-483-5p49315952
hsa-miR-48449325953
hsa-miR-485-3p49335954
hsa-miR-485-5p49345955
hsa-miR-486-3p49355956
hsa-miR-486-5p49365957
hsa-miR-487a49375958
hsa-miR-487b49385959
hsa-miR-488-3p49395960
hsa-miR-488-5p49405961
hsa-miR-48949415962
hsa-miR-490-3p49425963
hsa-miR-490-5p49435964
hsa-miR-491-3p49445965
hsa-miR-491-5p49455966
hsa-miR-49249465967
hsa-miR-493-3p49475968
hsa-miR-493-5p49485969
hsa-miR-49449495970
hsa-miR-495-3p49505971
hsa-miR-495-5p49515972
hsa-miR-49649525973
hsa-miR-497-3p49535974
hsa-miR-497-5p49545975
hsa-miR-49849555976
hsa-miR-4999-3p49565977
hsa-miR-4999-5p49575978
hsa-miR-499a-3p49585979
hsa-miR-499a-5p49595980
hsa-miR-499b-3p49605981
hsa-miR-499b-5p49615982
hsa-miR-5000-3p49625983
hsa-miR-5000-5p49635984
hsa-miR-5001-3p49645985
hsa-miR-5001-5p49655986
hsa-miR-5002-3p49665987
hsa-miR-5002-5p49675988
hsa-miR-5003-3p49685989
hsa-miR-5003-5p49695990
hsa-miR-5004-3p49705991
hsa-miR-5004-5p49715992
hsa-miR-5006-3p49725993
hsa-miR-5006-5p49735994
hsa-miR-5007-3p49745995
hsa-miR-5007-5p49755996
hsa-miR-5008-3p49765997
hsa-miR-5008-5p49775998
hsa-miR-5009-3p49785999
hsa-miR-5009-5p49796000
hsa-miR-500a-3p49806001
hsa-miR-500a-5p49816002
hsa-miR-500b49826003
hsa-miR-5010-3p49836004
hsa-miR-5010-5p49846005
hsa-miR-5011-3p49856006
hsa-miR-5011-5p49866007
hsa-miR-501-3p49876008
hsa-miR-501-5p49886009
hsa-miR-502-3p49896010
hsa-miR-502-5p49906011
hsa-miR-503-3p49916012
hsa-miR-503-5p49926013
hsa-miR-50449936014
hsa-miR-504749946015
hsa-miR-505-3p49956016
hsa-miR-505-5p49966017
hsa-miR-506-3p49976018
hsa-miR-506-5p49986019
hsa-miR-50749996020
hsa-miR-508-3p50006021
hsa-miR-508-5p50016022
hsa-miR-508750026023
hsa-miR-508850036024
hsa-miR-5089-3p50046025
hsa-miR-5089-5p50056026
hsa-miR-509050066027
hsa-miR-509150076028
hsa-miR-509250086029
hsa-miR-509350096030
hsa-miR-509-3-5p50106031
hsa-miR-509-3p50116032
hsa-miR-509450126033
hsa-miR-509550136034
hsa-miR-509-5p50146035
hsa-miR-509650156036
hsa-miR-51050166037
hsa-miR-510050176038
hsa-miR-51150186039
hsa-miR-512-3p50196040
hsa-miR-512-5p50206041
hsa-miR-513a-3p50216042
hsa-miR-513a-5p50226043
hsa-miR-513b50236044
hsa-miR-513c-3p50246045
hsa-miR-513c-5p50256046
hsa-miR-514a-3p50266047
hsa-miR-514a-5p50276048
hsa-miR-514b-3p50286049
hsa-miR-514b-5p50296050
hsa-miR-515-3p50306051
hsa-miR-515-5p50316052
hsa-miR-516a-3p50326053
hsa-miR-516a-5p50336054
hsa-miR-516b-3p50346055
hsa-miR-516b-5p50356056
hsa-miR-517-5p50366057
hsa-miR-517a-3p50376058
hsa-miR-517b-3p50386059
hsa-miR-517c-3p50396060
hsa-miR-518650406061
hsa-miR-5187-3p50416062
hsa-miR-5187-5p50426063
hsa-miR-518850436064
hsa-miR-518950446065
hsa-miR-518a-3p50456066
hsa-miR-518a-5p50466067
hsa-miR-518b50476068
hsa-miR-518c-3p50486069
hsa-miR-518c-5p50496070
hsa-miR-518d-3p50506071
hsa-miR-518d-5p50516072
hsa-miR-518e-3p50526073
hsa-miR-518e-5p50536074
hsa-miR-518f-3p50546075
hsa-miR-518f-5p50556076
hsa-miR-519050566077
hsa-miR-519150576078
hsa-miR-519250586079
hsa-miR-519350596080
hsa-miR-519450606081
hsa-miR-5195-3p50616082
hsa-miR-5195-5p50626083
hsa-miR-5196-3p50636084
hsa-miR-5196-5p50646085
hsa-miR-5197-3p50656086
hsa-miR-5197-5p50666087
hsa-miR-519a-3p50676088
hsa-miR-519a-5p50686089
hsa-miR-519b-3p50696090
hsa-miR-519b-5p50706091
hsa-miR-519c-3p50716092
hsa-miR-519c-5p50726093
hsa-miR-519d50736094
hsa-miR-519e-3p50746095
hsa-miR-519e-5p50756096
hsa-miR-520a-3p50766097
hsa-miR-520a-5p50776098
hsa-miR-520b50786099
hsa-miR-520c-3p50796100
hsa-miR-520c-5p50806101
hsa-miR-520d-3p50816102
hsa-miR-520d-5p50826103
hsa-miR-520e50836104
hsa-miR-520f50846105
hsa-miR-520g50856106
hsa-miR-520h50866107
hsa-miR-52150876108
hsa-miR-522-3p50886109
hsa-miR-522-5p50896110
hsa-miR-523-3p50906111
hsa-miR-523-5p50916112
hsa-miR-524-3p50926113
hsa-miR-524-5p50936114
hsa-miR-525-3p50946115
hsa-miR-525-5p50956116
hsa-miR-526a50966117
hsa-miR-526b-3p50976118
hsa-miR-526b-5p50986119
hsa-miR-52750996120
hsa-miR-532-3p51006121
hsa-miR-532-5p51016122
hsa-miR-539-3p51026123
hsa-miR-539-5p51036124
hsa-miR-541-3p51046125
hsa-miR-541-5p51056126
hsa-miR-542-3p51066127
hsa-miR-542-5p51076128
hsa-miR-54351086129
hsa-miR-544a51096130
hsa-miR-544b51106131
hsa-miR-545-3p51116132
hsa-miR-545-5p51126133
hsa-miR-54851136134
hsa-miR-548-3p51146135
hsa-miR-548-5p51156136
hsa-miR-548a51166137
hsa-miR-548a-3p51176138
hsa-miR-548a-5p51186139
hsa-miR-548aa51196140
hsa-miR-548ab51206141
hsa-miR-548ac51216142
hsa-miR-548ad51226143
hsa-miR-548ae51236144
hsa-miR-548ag51246145
hsa-miR-548ah-3p51256146
hsa-miR-548ah-5p51266147
hsa-miR-548ai51276148
hsa-miR-548aj-3p51286149
hsa-miR-548aj-5p51296150
hsa-miR-548ak51306151
hsa-miR-548al51316152
hsa-miR-548am-3p51326153
hsa-miR-548am-5p51336154
hsa-miR-548an51346155
hsa-miR-548ao-3p51356156
hsa-miR-548ao-5p51366157
hsa-miR-548ap-3p51376158
hsa-miR-548ap-5p51386159
hsa-miR-548aq-3p51396160
hsa-miR-548aq-5p51406161
hsa-miR-548ar-3p51416162
hsa-miR-548ar-5p51426163
hsa-miR-548as-3p51436164
hsa-miR-548as-5p51446165
hsa-miR-548at-3p51456166
hsa-miR-548at-5p51466167
hsa-miR-548au-3p51476168
hsa-miR-548au-5p51486169
hsa-miR-548av-3p51496170
hsa-miR-548av-5p51506171
hsa-miR-548aw51516172
hsa-miR-548ay-3p51526173
hsa-miR-548ay-5p51536174
hsa-miR-548az-3p51546175
hsa-miR-548az-5p51556176
hsa-miR-548b-3p51566177
hsa-miR-548b-5p51576178
hsa-miR-548c-3p51586179
hsa-miR-548c-5p51596180
hsa-miR-548d-3p51606181
hsa-miR-548d-5p51616182
hsa-miR-548e51626183
hsa-miR-548f51636184
hsa-miR-548g-3p51646185
hsa-miR-548g-5p51656186
hsa-miR-548h-3p51666187
hsa-miR-548h-5p51676188
hsa-miR-548i51686189
hsa-miR-548j51696190
hsa-miR-548k51706191
hsa-miR-548l51716192
hsa-miR-548m51726193
hsa-miR-548n51736194
hsa-miR-548o-3p51746195
hsa-miR-548o-5p51756196
hsa-miR-548p51766197
hsa-miR-548q51776198
hsa-miR-548s51786199
hsa-miR-548t-3p51796200
hsa-miR-548t-5p51806201
hsa-miR-548u51816202
hsa-miR-548w51826203
hsa-miR-548y51836204
hsa-miR-548z51846205
hsa-miR-549a51856206
hsa-miR-550a-3-5p51866207
hsa-miR-550a-3p51876208
hsa-miR-550a-5p51886209
hsa-miR-550b-2-5p51896210
hsa-miR-550b-3p51906211
hsa-miR-551a51916212
hsa-miR-551b-3p51926213
hsa-miR-551b-5p51936214
hsa-miR-55251946215
hsa-miR-55351956216
hsa-miR-55451966217
hsa-miR-55551976218
hsa-miR-556-3p51986219
hsa-miR-556-5p51996220
hsa-miR-55752006221
hsa-miR-5571-3p52016222
hsa-miR-5571-5p52026223
hsa-miR-557252036224
hsa-miR-5579-3p52046225
hsa-miR-5579-5p52056226
hsa-miR-55852066227
hsa-miR-5580-3p52076228
hsa-miR-5580-5p52086229
hsa-miR-5581-3p52096230
hsa-miR-5581-5p52106231
hsa-miR-5582-3p52116232
hsa-miR-5582-5p52126233
hsa-miR-5583-3p52136234
hsa-miR-5583-5p52146235
hsa-miR-5584-3p52156236
hsa-miR-5584-5p52166237
hsa-miR-5585-3p52176238
hsa-miR-5585-5p52186239
hsa-miR-5586-3p52196240
hsa-miR-5586-5p52206241
hsa-miR-5587-3p52216242
hsa-miR-5587-5p52226243
hsa-miR-5588-3p52236244
hsa-miR-5588-5p52246245
hsa-miR-5589-3p52256246
hsa-miR-5589-5p52266247
hsa-miR-55952276248
hsa-miR-5590-3p52286249
hsa-miR-5590-5p52296250
hsa-miR-5591-3p52306251
hsa-miR-5591-5p52316252
hsa-miR-561-3p52326253
hsa-miR-561-5p52336254
hsa-miR-56252346255
hsa-miR-56352356256
hsa-miR-56452366257
hsa-miR-56652376258
hsa-miR-56752386259
hsa-miR-56852396260
hsa-miR-568052406261
hsa-miR-5681a52416262
hsa-miR-5681b52426263
hsa-miR-568252436264
hsa-miR-568352446265
hsa-miR-568452456266
hsa-miR-568552466267
hsa-miR-568652476268
hsa-miR-568752486269
hsa-miR-568852496270
hsa-miR-568952506271
hsa-miR-56952516272
hsa-miR-569052526273
hsa-miR-569152536274
hsa-miR-5692a52546275
hsa-miR-5692b52556276
hsa-miR-5692c52566277
hsa-miR-569352576278
hsa-miR-569452586279
hsa-miR-569552596280
hsa-miR-569652606281
hsa-miR-569752616282
hsa-miR-569852626283
hsa-miR-569952636284
hsa-miR-570052646285
hsa-miR-570152656286
hsa-miR-570252666287
hsa-miR-570352676288
hsa-miR-570-3p52686289
hsa-miR-570452696290
hsa-miR-570552706291
hsa-miR-570-5p52716292
hsa-miR-570652726293
hsa-miR-570752736294
hsa-miR-570852746295
hsa-miR-57152756296
hsa-miR-57252766297
hsa-miR-57352776298
hsa-miR-573952786299
hsa-miR-574-3p52796300
hsa-miR-574-5p52806301
hsa-miR-57552816302
hsa-miR-576-3p52826303
hsa-miR-576-5p52836304
hsa-miR-57752846305
hsa-miR-57852856306
hsa-miR-578752866307
hsa-miR-57952876308
hsa-miR-58052886309
hsa-miR-58152896310
hsa-miR-582-3p52906311
hsa-miR-582-5p52916312
hsa-miR-58352926313
hsa-miR-584-3p52936314
hsa-miR-584-5p52946315
hsa-miR-58552956316
hsa-miR-58652966317
hsa-miR-58752976318
hsa-miR-58852986319
hsa-miR-589-3p52996320
hsa-miR-589-5p53006321
hsa-miR-590-3p53016322
hsa-miR-590-5p53026323
hsa-miR-59153036324
hsa-miR-59253046325
hsa-miR-593-3p53056326
hsa-miR-593-5p53066327
hsa-miR-59553076328
hsa-miR-59653086329
hsa-miR-59753096330
hsa-miR-59853106331
hsa-miR-59953116332
hsa-miR-60053126333
hsa-miR-60153136334
hsa-miR-60253146335
hsa-miR-60353156336
hsa-miR-60453166337
hsa-miR-60553176338
hsa-miR-60653186339
hsa-miR-606853196340
hsa-miR-606953206341
hsa-miR-60753216342
hsa-miR-607053226343
hsa-miR-607153236344
hsa-miR-607253246345
hsa-miR-607353256346
hsa-miR-607453266347
hsa-miR-607553276348
hsa-miR-607653286349
hsa-miR-607753296350
hsa-miR-607853306351
hsa-miR-607953316352
hsa-miR-60853326353
hsa-miR-608053336354
hsa-miR-608153346355
hsa-miR-608253356356
hsa-miR-608353366357
hsa-miR-608453376358
hsa-miR-608553386359
hsa-miR-608653396360
hsa-miR-608753406361
hsa-miR-608853416362
hsa-miR-608953426363
hsa-miR-60953436364
hsa-miR-609053446365
hsa-miR-61053456366
hsa-miR-61153466367
hsa-miR-61253476368
hsa-miR-612453486369
hsa-miR-612553496370
hsa-miR-612653506371
hsa-miR-612753516372
hsa-miR-612853526373
hsa-miR-612953536374
hsa-miR-61353546375
hsa-miR-613053556376
hsa-miR-613153566377
hsa-miR-613253576378
hsa-miR-613353586379
hsa-miR-613453596380
hsa-miR-61453606381
hsa-miR-615-3p53616382
hsa-miR-615-5p53626383
hsa-miR-616-3p53636384
hsa-miR-616553646385
hsa-miR-616-5p53656386
hsa-miR-61753666387
hsa-miR-61853676388
hsa-miR-61953686389
hsa-miR-62053696390
hsa-miR-62153706391
hsa-miR-62253716392
hsa-miR-62353726393
hsa-miR-624-3p53736394
hsa-miR-624-5p53746395
hsa-miR-625-3p53756396
hsa-miR-625-5p53766397
hsa-miR-62653776398
hsa-miR-62753786399
hsa-miR-628-3p53796400
hsa-miR-628-5p53806401
hsa-miR-629-3p53816402
hsa-miR-629-5p53826403
hsa-miR-63053836404
hsa-miR-63153846405
hsa-miR-63253856406
hsa-miR-63353866407
hsa-miR-63453876408
hsa-miR-63553886409
hsa-miR-63653896410
hsa-miR-63753906411
hsa-miR-63853916412
hsa-miR-63953926413
hsa-miR-64053936414
hsa-miR-64153946415
hsa-miR-642a-3p53956416
hsa-miR-642a-5p53966417
hsa-miR-642b-3p53976418
hsa-miR-642b-5p53986419
hsa-miR-64353996420
hsa-miR-644a54006421
hsa-miR-64554016422
hsa-miR-64654026423
hsa-miR-64754036424
hsa-miR-64854046425
hsa-miR-64954056426
hsa-miR-6499-3p54066427
hsa-miR-6499-5p54076428
hsa-miR-65054086429
hsa-miR-6500-3p54096430
hsa-miR-6500-5p54106431
hsa-miR-6501-3p54116432
hsa-miR-6501-5p54126433
hsa-miR-6502-3p54136434
hsa-miR-6502-5p54146435
hsa-miR-6503-3p54156436
hsa-miR-6503-5p54166437
hsa-miR-6504-3p54176438
hsa-miR-6504-5p54186439
hsa-miR-6505-3p54196440
hsa-miR-6505-5p54206441
hsa-miR-6506-3p54216442
hsa-miR-6506-5p54226443
hsa-miR-6507-3p54236444
hsa-miR-6507-5p54246445
hsa-miR-6508-3p54256446
hsa-miR-6508-5p54266447
hsa-miR-6509-3p54276448
hsa-miR-6509-5p54286449
hsa-miR-65154296450
hsa-miR-6510-3p54306451
hsa-miR-6510-5p54316452
hsa-miR-6511a-3p54326453
hsa-miR-6511a-5p54336454
hsa-miR-6511b-3p54346455
hsa-miR-6511b-5p54356456
hsa-miR-6512-3p54366457
hsa-miR-6512-5p54376458
hsa-miR-6513-3p54386459
hsa-miR-6513-5p54396460
hsa-miR-6514-3p54406461
hsa-miR-6514-5p54416462
hsa-miR-6515-3p54426463
hsa-miR-6515-5p54436464
hsa-miR-652-3p54446465
hsa-miR-652-5p54456466
hsa-miR-65354466467
hsa-miR-654-3p54476468
hsa-miR-654-5p54486469
hsa-miR-65554496470
hsa-miR-65654506471
hsa-miR-65754516472
hsa-miR-65854526473
hsa-miR-659-3p54536474
hsa-miR-659-5p54546475
hsa-miR-660-3p54556476
hsa-miR-660-5p54566477
hsa-miR-66154576478
hsa-miR-66254586479
hsa-miR-663a54596480
hsa-miR-663b54606481
hsa-miR-664a-3p54616482
hsa-miR-664a-5p54626483
hsa-miR-664b-3p54636484
hsa-miR-664b-5p54646485
hsa-miR-66554656486
hsa-miR-66854666487
hsa-miR-67054676488
hsa-miR-671-3p54686489
hsa-miR-6715a-3p54696490
hsa-miR-6715b-3p54706491
hsa-miR-6715b-5p54716492
hsa-miR-671-5p54726493
hsa-miR-6716-3p54736494
hsa-miR-6716-5p54746495
hsa-miR-6717-5p54756496
hsa-miR-6718-5p54766497
hsa-miR-6719-3p54776498
hsa-miR-6720-3p54786499
hsa-miR-6721-5p54796500
hsa-miR-6722-3p54806501
hsa-miR-6722-5p54816502
hsa-miR-6723-5p54826503
hsa-miR-6724-5p54836504
hsa-miR-675-3p54846505
hsa-miR-675-5p54856506
hsa-miR-676-3p54866507
hsa-miR-676-5p54876508
hsa-miR-708-3p54886509
hsa-miR-708-5p54896510
hsa-miR-71154906511
hsa-miR-7-1-3p54916512
hsa-miR-71854926513
hsa-miR-7-2-3p54936514
hsa-miR-744-3p54946515
hsa-miR-744-5p54956516
hsa-miR-758-3p54966517
hsa-miR-758-5p54976518
hsa-miR-75954986519
hsa-miR-7-5p54996520
hsa-miR-76055006521
hsa-miR-76155016522
hsa-miR-76255026523
hsa-miR-76455036524
hsa-miR-76555046525
hsa-miR-766-3p55056526
hsa-miR-766-5p55066527
hsa-miR-767-3p55076528
hsa-miR-767-5p55086529
hsa-miR-769-3p55096530
hsa-miR-769-5p55106531
hsa-miR-770-5p55116532
hsa-miR-80255126533
hsa-miR-873-3p55136534
hsa-miR-873-5p55146535
hsa-miR-87455156536
hsa-miR-875-3p55166537
hsa-miR-875-5p55176538
hsa-miR-876-3p55186539
hsa-miR-876-5p55196540
hsa-miR-877-3p55206541
hsa-miR-877-5p55216542
hsa-miR-885-3p55226543
hsa-miR-885-5p55236544
hsa-miR-88755246545
hsa-miR-888-3p55256546
hsa-miR-888-5p55266547
hsa-miR-88955276548
hsa-miR-89055286549
hsa-miR-891a55296550
hsa-miR-891b55306551
hsa-miR-892a55316552
hsa-miR-892b55326553
hsa-miR-892c-3p55336554
hsa-miR-892c-5p55346555
hsa-miR-92055356556
hsa-miR-92155366557
hsa-miR-92255376558
hsa-miR-92455386559
hsa-miR-92a-1-5p55396560
hsa-miR-92a-2-5p55406561
hsa-miR-92a-3p55416562
hsa-miR-92b-3p55426563
hsa-miR-92b-5p55436564
hsa-miR-93355446565
hsa-miR-93-3p55456566
hsa-miR-93455466567
hsa-miR-93555476568
hsa-miR-93-5p55486569
hsa-miR-93655496570
hsa-miR-937-3p55506571
hsa-miR-937-5p55516572
hsa-miR-93855526573
hsa-miR-939-3p55536574
hsa-miR-939-5p55546575
hsa-miR-9-3p55556576
hsa-miR-94055566577
hsa-miR-94155576578
hsa-miR-94255586579
hsa-miR-94355596580
hsa-miR-94455606581
hsa-miR-9555616582
hsa-miR-9-5p55626583
hsa-miR-96-3p55636584
hsa-miR-96-5p55646585
hsa-miR-98-3p55656586
hsa-miR-98-5p55666587
hsa-miR-99a-3p55676588
hsa-miR-99a-5p55686589
hsa-miR-99b-3p55696590
hsa-miR-99b-5p55706591

As shown in Table 10, microRNAs are differentially expressed in different tissues and cells, and often associated with different types of diseases (e.g. cancer cells). The decision of removal or insertion of microRNA binding sites, or any combination, is dependent on microRNA expression patterns and their profilings in cancer cells. In Table 10, “HCC” represents hepatocellular carcinoma, “ALL” stands for acute lymphoblastic leukemia, “RCC” stands for renal cell carcinoma, “CLL” stands for chromic lymphocytic leukemia and “MALT” stands for mucosa-associated lymphoid tissue.

TABLE 10
mirs, tissues/cell expression and diseases
BS
mirSEQAssociatedBiological
microRNASEQ IDIDTissues/cellsDiseaseFunction
hsa-let-7a-2-3p25083529Embryonic steminflammatory,tumor
cells, lung, myeloidvarious cancerssuppressor
cells(lung, cervical,
breast, pancreatic,
etc)
hsa-let-7a-3p25093530Embryonic steminflammatory,tumor
cells, lungvarious cancerssuppressor
(lung, cervical,
breast, pancreatic,
etc)
hsa-let-7a-5p25103531Embryonic steminflammatory,tumor
cells, lungvarious cancerssuppressor
(lung, cervical,
breast, pancreatic,
etc)
hsa-let-7b-3p25113532epithelial cells,lung cancer,tumor
endothelial cellscolorectal cancer,angiogenesis
(vascular)cervical cancer,
inflammation and
immune response
after infection
hsa-let-7b-5p25123533epithelial cells,cervical cancer,tumor
endothelial cellsinflammation andangiogenesis
(vascular)immune response
after infection
hsa-let-7c25133534dendritic cellsvarious cacnerstumor
(cervical,suppressor,
pancreatic,apoptosis
lung,
esopphageal, etc)
hsa-let-7d-3p25143535embryonic stemassociated withtumor
cellsvarious cancersuppressor
cells
hsa-let-7d-5p25153536embryonic stemassociated withtumor
cellsvarious cancersuppressor
cells
hsa-let-7e-3p25163537immune cellsvarious cancertumor
cells,suppressor
autoimmunity,
endotoxin
tolerance
hsa-let-7e-5p25173538immune cellsvarious cancertumor
cellssuppressor
hsa-let-7f-1-3p25183539immune cells (Tvarious cancertumor
cells)cellssuppressor
hsa-let-7f-2-3p25193540immune cells (Tvarious cancertumor
cells)cellssuppressor
hsa-let-7f-5p25203541immune cells (TVarious cancertumor
cells)cellssuppressor
hsa-let-7g-3p25213542hematopoietic cells,various cancertumor
adipose, smoothcells (lung, breast,suppressor
muscle cellsetc)
hsa-let-7g-5p25223543hematopoietic cells,various cancertumor
adipose, smoothcells (lung, breast,suppressor
muscle cellsetc)
hsa-let-7i-3p25233544immune cellschronictumor
lymphocytesuppressor
leukimia
hsa-let-7i-5p25243545immune cellschronictumor
lymphocytesuppressor
leukimia
hsa-miR-125253546muscle, heartangiogenesis,
cell
proliferation (myogenesis)
hsa-miR-100-3p25263547hematopoietic cells,gastric cancer,tumor
endothelial cellspancreatic cancerangiogenesis
hsa-miR-100-5p25273548hematopoietic cells,gastric cancer,tumor
endothelial cellspancreatic cancerangiogenesis
hsa-miR-101-3p25283549endothelial cellsvarious cancersangiogenesis
(breast, non-small
cell lung, colon,
gastric,
pancreatic,
bladder, etc);
lupus
erythematosus
hsa-miR-101-5p25293550endothelial cellsvarious cancersangiogenesis
(breast, non-small
cell lung, colon,
gastric,
pancreatic,
bladder, etc);
lupus
erythematosus
hsa-miR-103a-2-5p25303551embryonic stemvarious cancersoncogene, cell
cells, many(endometrial,growth
tissues/cellsneuroblastoma,
colorectal, breast,
liver, etc)
hsa-miR-103a-3p25313552embryonic stemvarious cancersoncogene, cell
cells, many(endometrial,growth
tissues/cellsneuroblastoma,
colorectal, breast,
liver, etc)
hsa-miR-103b25323553Many tissues/cellsvarious cancersoncogene, cell
(endometrial,growth
neuroblastoma,
colorectal, breast,
liver, etc)
hsa-miR-105-3p25333554pancreatic cells
hsa-miR-105-5p25343555pancreatic cells
hsa-miR-106a-3p25353556osteogenic cellsosteocarcoma,cell
other cancersdifferentiation
hsa-miR-106a-5p25363557osteogenic cellsosteocarcoma,cell
other cancersdifferentiation
hsa-miR-106b-3p25373558embryonic stemvarious cancersoncogene
cells(non-small lung
cancer,
gastric cancer,
HCC, gliomas,
etc)
hsa-miR-106b-5p25383559embryonic stemvarious cancersoncogene
cells(non-small lung
cancer,
gastric cancer,
HCC, gliomas,
etc)
hsa-miR-10725393560many tissues, brainbreast cancer,
hepatocytes/liverpituitary
adenoma,
obesity/diabetes
hsa-miR-10a-3p25403561hematopoeitic cellsacute myeoidoncogene, cell
leukemiagrowth
hsa-miR-10a-5p25413562hematopoeitic cellsacute myeoidoncogene, cell
leukemiagrowth
hsa-miR-10b-3p25423563multiple tissues andvarious cancersoncogene
cells(breast, ovarian,
glioblastoma,
pancreatc ductal
adenocarcinoma,
gastric, etc)
hsa-miR-10b-5p25433564multiple tissues andvarious cancersoncogene
cells(breast, ovarian,
glioblastoma,
pancreatc ductal
adenocarcinoma,
gastric, etc)
hsa-miR-1178-3p25443565osteocarcoma
hsa-miR-1178-5p25453566osteocarcoma
hsa-miR-117925463567osteocarcoma
hsa-miR-118025473568discovered in
sarcoma, no
expression data
hsa-miR-118125483569downregulated in
ovarian cancer
cells,
associated with
HCV infection in
hepatocytes
hsa-miR-118225493570placenta
hsa-miR-118325503571associated with
rectal cancer
hsa-miR-118425513572Hematopoietic cellsdownregulated in
oral leukoplakia
(OLK)
hsa-miR-1185-1-3p25523573placenta
hsa-miR-1185-2-3p25533574placenta
hsa-miR-1185-5p25543575placenta
hsa-miR-119325553576melanoma
hsa-miR-119725563577neublastoma
hsa-miR-120025573578chronic
lynphocytic
leukemia
hsa-miR-120225583579chronic
lynphocytic
leukemia,
downregulated in
ovarian cancer
cells
hsa-miR-120325593580in the
chromosome
8q24 region,
cancer cells
hsa-miR-120425603581in the
chromosome
8q24 region,
cancer cells
hsa-miR-120525613582in the
chromosome
8q24 region,
cancer cells
hsa-miR-120625623583in the
chromosome
8q24 region,
cancer cells
hsa-miR-1207-3p25633584in the
chromosome
8q24 region,
cancer cells
hsa-miR-1207-5p25643585in the
chromosome
8q24 region,
cancer cells
hsa-miR-120825653586in the
chromosome
8q24 region,
cancer cells
hsa-miR-122-3p25663587kidney,Renal Celllipid
liver/hepatocytesCarcinomametabolism
(RCC), cancer
cells
hsa-miR-1224-3p25673588Lupus nephritis
hsa-miR-1224-5p25683589rectal cancer
hsa-miR-1225-3p25693590adrenal
pheochromocytomas;
upregulated in
MITF
KnockDown
melanocytes
hsa-miR-1225-5p25703591prostate cancer
hsa-miR-122-5p25713592liver/hepatocytescancer cellslipid
metabolism
hsa-miR-1226-3p25723593discovered in a
mirtron screening
hsa-miR-1226-5p25733594discovered in a
mirtron screening
hsa-miR-1227-3p25743595cartilage/chondrocytes
hsa-miR-1227-5p25753596cartilage/chondrocytes
hsa-miR-1228-3p25763597liver (hepatocytes)Hepatocellularanti-apoptosis
carcinoma (HCC)
hsa-miR-1228-5p25773598liver (hepatocytes)Hepatocellularanti-apoptosis
carcinoma (HCC)
hsa-miR-1229-3p25783599discovered in a
mirtron screening
hsa-miR-1229-5p25793600discovered in a
mirtron screening
hsa-miR-123125803601HCC
hsa-miR-1233-1-5p25813602serum
hsa-miR-1233-3p25823603serum
hsa-miR-1234-3p25833604discovered in
embryonic stem
cell
hsa-miR-1234-5p25843605discovered in
embryonic stem
cell
hsa-miR-1236-3p25853606lymphatictarget to
endothelial cellsVEGFR-3
hsa-miR-1236-5p25863607lymphatictarget to
endothelial cellsVEGFR-3
hsa-miR-1237-3p25873608esophageal cell line
KYSE-150R
hsa-miR-1237-5p25883609esophageal cell line
KYSE-150R
hsa-miR-1238-3p25893610colorectal cancer
hsa-miR-1238-5p25903611colorectal cancer
hsa-miR-124325913612discovered in
embryonic stem
cells
hsa-miR-124-3p25923613brain, plasmagliomacell
(exosomal)differentiation
hsa-miR-124425933614discovered in
embryonic stem
cells
hsa-miR-1245a25943615discovered in
embryonic stem
cells
hsa-miR-1245b-3p25953616discovered in
embryonic stem
cells
hsa-miR-1245b-5p25963617discovered in
embryonic stem
cells
hsa-miR-124-5p25973618brain, Plasmaupregulated incell
(circulating)heart dysfunction,differentiation
glioma
hsa-miR-124625983619embryonic stem
cells, epithelial
cells
hsa-miR-1247-3p25993620embryoid body
cells
hsa-miR-1247-5p26003621embryoid body
cells
hsa-miR-124826013622component of
SnoRNAs
hsa-miR-124926023623liver (hepatocytes)
hsa-miR-125026033624oligodendrocytes
hsa-miR-125126043625discovered in
embryonic stem
cells
hsa-miR-125226053626discovered in
embryonic stem
cells
hsa-miR-125326063627discovered in
embryonic stem
cells
hsa-miR-125426073628embryonic stem
cells
hsa-miR-1255a26083629discovered in
embryonic stem
cells
hsa-miR-1255b-2-3p26093630discovered in
embryonic stem
cells
hsa-miR-1255b-5p26103631discovered in
embryonic stem
cells
hsa-miR-125626113632discovered inprostate cancer
embryonic stem
cells
hsa-miR-125726123633discovered inliposarcoma (soft
embryonic stemtissue sarcoma)
cells
hsa-miR-125826133634discovered inbreast cancer and
embryonic stemlung cancer
cells
hsa-miR-125a-3p26143635brain,various cancercell proliferation
hematopoietic cells(prostate, HCC,and
etc)differentiation
hsa-miR-125a-5p26153636brain,various cancercell proliferation
hematopoietic cells(prostate, HCC,and
etc)differentiation
hsa-miR-125b-1-3p26163637hematopoietic cellsvarious canceroncogene, cell
(monocytes),(prostate, HCC,differentiation
brain(neuron)etc)
hsa-miR-125b-2-3p26173638hematopoietic cellsvarious canceroncogene, cell
(monocytes),(prostate, HCC,differentiation
brain(neuron)etc)
hsa-miR-125b-5p26183639hematopoietic cells,various canceroncogene, cell
brain (neuron)(cutaneous T celldifferentiation
lymphoma,
prostate, HCC,
etc)
hsa-miR-1260a26193640periodontal tissue
hsa-miR-1260b26203641periodontal tissue
hsa-miR-126126213642embryonic stem
cells
hsa-miR-126226223643embryoid body
cells
hsa-miR-126326233644discovered in
embryonic stem
cells
hsa-miR-126-3p26243645endothelialB-lieage ALLangiogenesis
cells, lung
hsa-miR-126426253646discovered in
embryonic stem
cells
hsa-miR-126526263647discovered in
embryonic stem
cells
hsa-miR-126-5p26273648endothelialbreast cancer, B-angiogenesis
cells, lunglieage ALL
hsa-miR-126626283649embryonic stem
cells
hsa-miR-126726293650discovered in
embryonic stem
cells
hsa-miR-1268a26303651embryonic stem
cells
hsa-miR-1268b26313652embryonic stem
cells
hsa-miR-1269a26323653embryoid body
cells
hsa-miR-1269b26333654embryoid body
cells
hsa-miR-127026343655discovered in
embryonic stem
cells
hsa-miR-1271-3p26353656brainHepatocellularSuppress GPC-3
carcinoma (HCC)in HCC
hsa-miR-1271-5p26363657brainHepatocellularSuppress GPC-3
carcinoma (HCC)in HCC
hsa-miR-127226373658embryonic stem
cells
hsa-miR-1273a26383659discovered in
embryonic stem
cells
hsa-miR-1273c26393660colorectal cancer
hsa-miR-1273d26403661discovered in
embryonic stem
cells
hsa-miR-1273e26413662solid tumor cells
hsa-miR-1273f26423663cervical cancer
hsa-miR-1273g-3p26433664cervical cancer
hsa-miR-1273g-5p26443665cervical cancer
hsa-miR-127-3p26453666lung, placenta
hsa-miR-127526463667embryonic stemgastric carcinoma
cells
hsa-miR-127-5p26473668lung, placenta (islet)
hsa-miR-127626483669discovered in
embryonic stem
cells
hsa-miR-1277-3p26493670embryoid body
cells
hsa-miR-1277-5p26503671embryoid body
cells
hsa-miR-127826513672discovered in
embryonic stem
cells
hsa-miR-127926523673monocytes
hsa-miR-12826533674glioblast, brainB-lieage ALLtarget to
neurofibrominlin
neuron
hsa-miR-128126543675muscle invasive
bladder cancer
hsa-miR-128226553676discovered in
embryonic stem
cells
hsa-miR-128326563677placenta
hsa-miR-128426573678lung cancer
hsa-miR-1285-3p26583679various cancerinhibit P53
cellsexpression
hsa-miR-1285-5p26593680various cancerinhibit P53
cellsexpression
hsa-miR-128626603681smooth muscleesophageal cancer
hsa-miR-128726613682embryoid bodybreast cancer
cells
hsa-miR-128826623683discovered in
embryonic stem
cells
hsa-miR-128926633684multiple cell types
hsa-miR-129026643685embryoid bodygastric carcinoma
cells
hsa-miR-129126653686hepatocytescomponent of
SnoRNAs
hsa-miR-129-1-3p26663687multiple cell typesHCC cancer cells
hsa-miR-1292-3p26673688
hsa-miR-129-2-3p26683689multiple cell typesvarious cancer
cells
hsa-miR-1292-5p26693690
hsa-miR-129326703691discovered in
embryonic stem
cells
hsa-miR-129426713692discovered in
embryonic stem
cells
hsa-miR-1295a26723693tumor cells
(follicular
lymphoma)
hsa-miR-1295b-3p26733694tumor cells
(follicular
lymphoma)
hsa-miR-1295b-5p26743695tumor cells
(follicular
lymphoma)
hsa-miR-129-5p26753696liver (hepatocytes)HCC, thyroidcell death in
cancercancer cell
hsa-miR-129626763697breast cancer
hsa-miR-129726773698discovered in
embryonic stem
cells
hsa-miR-129826783699
hsa-miR-129926793700discovered in
embryonic stem
cells
hsa-miR-130126803701breast cancer
hsa-miR-130226813702
hsa-miR-130326823703hepatocytecolorectal cancer,
liver cancer
hsa-miR-1304-3p26833704dental
development
hsa-miR-1304-5p26843705dental
development
hsa-miR-130526853706discovered in
embryonic stem
cells
hsa-miR-1306-3p26863707discovered in
embryonic stem
cells
hsa-miR-1306-5p26873708discovered in
embryonic stem
cells
hsa-miR-1307-3p26883709discovered in
embryonic stem
cells
hsa-miR-1307-5p26893710discovered in
embryonic stem
cells
hsa-miR-130a-3p26903711lung, monocytes,various cancerspro-angiogenic
vascular endothelial(basal cell
cellscarcinoma,
HCC, ovarian,
etc), drug
resistance
hsa-miR-130a-5p26913712lung, monocytes,various cancerspro-angiogenic
vascular endothelial(basal cell
cellscarcinoma,
HCC, ovarian,
etc), drug
resistance
hsa-miR-130b-3p26923713Lung, epidermalvarious cancerscell
cells (keratinocytes)(gastric, rena cellproiferation/senescence
carcinoma)
hsa-miR-130b-5p26933714Lung, epidermalvarious cancerscell
cells (keratinocytes)(gastric, rena cellproiferation/senescence
carcinoma)
hsa-miR-132126943715neuroblastoma
hsa-miR-132226953716neuroblastoma
hsa-miR-132326963717placentaneuroblastoma
hsa-miR-132-3p26973718Brain (neuron),
immune cells
hsa-miR-132426983719neuroblastoma
hsa-miR-132-5p26993720brain (neuron),
immune cells
hsa-miR-133a27003721muscle, heart,heart failure,myogenesis
epithelial cellsesophageal cancer
(lung)
hsa-miR-133b27013722muscle, heart,heart failure,myogenesis
epithelial cellsesophageal cancer
(lung)
hsa-miR-13427023723lung (epithelial)non-samll cell
lung cancer,
pulmonary
embolism
hsa-miR-134327033724breast cancer cells
hsa-miR-135a-3p27043725brain, other tissuesvarious cancertumor
cells (lung, breast,suppressor
colorectal, HCC,
etc)
hsa-miR-135a-5p27053726brain, other tissuesvarious cancertumor
cells (lung, breast,suppressor
colorectal, HCC,
etc)
hsa-miR-135b-3p27063727brain, placenta,various cancers
other tissues(gastric,
mammary, neuroblastomas,
pancreatic, etc)
hsa-miR-135b-5p27073728brain, placenta,various cancers
other tissues(gastric,
mammary, neuroblastomas,
pancreatic, etc)
hsa-miR-136-3p27083729stem cells, placentagliomatumor
suppressor
hsa-miR-136-5p27093730stem cells, placentagliomatumor
suppressor
hsa-miR-13727103731brainvarious cancersinhibiting
(glioblastoma,cancer cell
breast, gastricproliferation and
etc), Alzheimer'smigration
disease
hsa-miR-138-1-3p27113732stem cells,arious cancercell
epidermalcells,proliferation/senescence
cells (keratinocytes)downregulated in
HCC
hsa-miR-138-2-3p27123733stem cellsarious cancer
cells,
downregulated in
HCC
hsa-miR-138-5p27133734stem cellsarious cancer
cells,
downregulated in
HCC
hsa-miR-139-3p27143735hematocytes, brainvarious cancerrepress cancer
cells (colorectal,metastasis
gastric, ovarian)
hsa-miR-139-5p27153736hematocytes, brainvarious cancerrepress cancer
cells (colorectal,metastasis
gastric, ovarian)
hsa-miR-140-3p27163737airway smoothVirus infection,
musclecancers
hsa-miR-140-5p27173738cartilagecsncers
(chondrocytes)
hsa-miR-141-3p27183739Many tissues/cellsvarious cancercell
cells (HCC,differentiation
prostate, kidney,
etc)
hsa-miR-141-5p27193740Many tissues/cellsvarious cancercell
cells (HCC,differentiation
prostate, kidney,
etc)
hsa-miR-142-3p27203741meyloid cells,immune
hematopoiesis,response
APC cells
hsa-miR-142-5p27213742meyloid cells,immune
hematopoiesis,response
APC cells
hsa-miR-143-3p27223743vascular smoothpre-B-cell acute
musclelymphocytic
leukemia, virus
infection
hsa-miR-143-5p27233744vascular smoothvirus infection
muscle, T-cells
hsa-miR-144-3p27243745erythroidvarious cancerscell
(lung, colorectal,differentiation
etc)
hsa-miR-144-5p27253746erythroidvarious cancerscell
(lung, colorectal,differentiation
etc)
hsa-miR-145-3p27263747kidney, cartilage,T-cell lupustumor
vascular smoothsuppressor
muscle
hsa-miR-145-5p27273748kidney, cartilage,T-cell lupustumor
vascular smoothsuppressor
muscle
hsa-miR-146827283749lung cancer
hsa-miR-146927293750tumor
cell (follicular
lymphoma), rectal
cancer
hsa-miR-146a-3p27303751immune cells,various cancers,
hematopoiesisendotoxin
tolerance
hsa-miR-146a-5p27313752immune cells,various cancers,
hematopoiesisendotoxin
tolerance
hsa-miR-146b-3p27323753immune cellsvarious cancers
hsa-miR-146b-5p27333754Embryonic stemvarious cancerstumor invation,
cells(glioma)migration
hsa-miR-147027343755
hsa-miR-147127353756tumor
cell (follicular
lymphoma), rectal
cancer
hsa-miR-147a27363757Macrophageinflammatory
response
hsa-miR-147b27373758Macrophageinflammatory
response
hsa-miR-148a-3p27383759hematopoietic cellsCLL, T-lineage
ALL
hsa-miR-148a-5p27393760hematopoietic cellsCLL, T-lineage
ALL
hsa-miR-148b-3p27403761neuron
hsa-miR-148b-5p27413762neuron
hsa-miR-149-3p27423763heart, brainvarious cancers
(glioma,
colorectal, gastric,
etc)
hsa-miR-149-5p27433764heart, brainvarious cancers
(glioma,
colorectal, gastric,
etc)
hsa-miR-150-3p27443765hematopoietic cellscirculating plasma
(lymphoid)(acute myeloid
leukemia)
hsa-miR-150-5p27453766hematopoietic cellscirculating plasma
(lymphoid)(acute myeloid
leukemia)
hsa-miR-151a-3p27463767neuron, fetal liver
hsa-miR-151a-5p27473768neuron, fetal liver
hsa-miR-151b27483769immune cells (B-
cells)
hsa-miR-15227493770liver
hsa-miR-15327503771brain
hsa-miR-153727513772
hsa-miR-153827523773bloodCancer cells
hsa-miR-153927533774esophageal cell line
KYSE-150R
hsa-miR-154-3p27543775embryonic stem
cells
hsa-miR-154-5p27553776embryonic stem
cells
hsa-miR-155-3p27563777T/B cells,various cancers
monocytes, breast(CLL, B cell
lymphoma,
breast, lung,
ovarian, cervical,
colorectal,
prostate)
hsa-miR-155-5p27573778T/B cells,various cancers
monocytes, breast(CLL, B cell
lymphoma,
breast, lung,
ovarian, cervical,
colorectal,
prostate)
hsa-miR-158727583779identified in B-cells
hsa-miR-15a-3p27593780blood, lymphocyte,cell cycle,
hematopoieticproliferation
tissues (spleen)
hsa-miR-15a-5p27603781blood, lymphocyte,cell cycle,
hematopoieticproliferation
tissues (spleen)
hsa-miR-15b-3p27613782blood, lymphocyte,cell cycle,
hematopoieticproliferation
tissues (spleen)
hsa-miR-15b-5p27623783blood, lymphocyte,cell cycle,
hematopoieticproliferation
tissues (spleen)
hsa-miR-16-1-3p27633784embryonic stem
cells, blood,
hematopoietic
tissues (spleen)
hsa-miR-16-2-3p27643785blood, lymphocyte,
hematopoietic
tissues (spleen)
hsa-miR-16-5p27653786Many tissues, blood
hsa-miR-17-3p27663787embryonic stemtumor
cells, endothelialangiogenesis
cells,
hsa-miR-17-5p27673788endothelial cells,tumor
kidney, breast;angiogenesis
hsa-miR-181a-2-3p27683789glioblast, stem cells
hsa-miR-181a-3p27693790glioblast, myeloid
cells, Embryonic
stem cells
hsa-miR-181a-5p27703791glioblast, myeloid
cells, Embryonic
stem cells
hsa-miR-181b-3p27713792glioblast,cell
Embryonic stemproiferation/senescence
cells, epidermal
(keratinocytes)
hsa-miR-181b-5p27723793glioblast,cell
Embryonic stemproiferation/senescence
cells, epidermal
(keratinocytes)
hsa-miR-181c-3p27733794brain, stemvariou cance cellscell
cells/progenitor(gliobasltoma,differentiation
basal cell
carcinoma,
prostate)
hsa-miR-181c-5p27743795brain, stemvariou cance cellscell
cells/progenitor(gliobasltoma,differentiation
basal cell
carcinoma,
prostate)
hsa-miR-181d27753796glia cells
hsa-miR-182-3p27763797immune cellsautoimmuneimmune
response
hsa-miR-182527773798discovered in a
MiRDeep screening
hsa-miR-182-5p27783799lung, immune cellsautoimmuneimmune
response
hsa-miR-182727793800small cell lung
cancer
hsa-miR-183-3p27803801brain
hsa-miR-183-5p27813802brain
hsa-miR-18427823803blood, tongue,
pancreas (islet)
hsa-miR-185-3p27833804
hsa-miR-185-5p27843805
hsa-miR-186-3p27853806osteoblasts, heartvarious cancer
cells
hsa-miR-186-5p27863807osteoblasts, heartvarious cancer
cells
hsa-miR-187-3p27873808thyroid tumor
hsa-miR-187-5p27883809thyroid tumor
hsa-miR-188-3p27893810irway smooth
muscle, central
nervous system
hsa-miR-188-5p27903811irway smooth
muscle, central
nervous system
hsa-miR-18a-3p27913812endothelial cells,
lung
hsa-miR-18a-5p27923813endothelial cells,
lung
hsa-miR-18b-3p27933814lung
hsa-miR-18b-5p27943815lung
hsa-miR-190827953816breast cancer
hsa-miR-1909-3p27963817rectal cancer
hsa-miR-1909-5p27973818rectal cancer
hsa-miR-190a27983819brain
hsa-miR-190b27993820brain
hsa-miR-191028003821embryonic stem
cells
hsa-miR-1911-3p28013822embryonic stem
cells, neural
precursor
hsa-miR-1911-5p28023823embryonic stem
cells, neural
precursor
hsa-miR-191228033824embryonic stem
cells, neural
precursor
hsa-miR-191328043825embryonic stem
cells
hsa-miR-191-3p28053826chroninc
lymphocyte
leukimia, B-
lieage ALL
hsa-miR-1914-3p28063827embryonic stem
cells
hsa-miR-1914-5p28073828embryonic stem
cells
hsa-miR-1915-3p28083829embryonic stem
cells
hsa-miR-1915-5p28093830embryonic stem
cells
hsa-miR-191-5p28103831chroninc
lymphocyte
leukimia, B-
lieage ALL
hsa-miR-192-3p28113832kidney
hsa-miR-192-5p28123833kidney
hsa-miR-193a-3p28133834many tissues/cellsvarious cancertumor
cells (lung,suppressor,
osteoblastoma,proliferation
ALL, follicular
lymphoma, etc)
hsa-miR-193a-5p28143835many tissues/cellsvarious cancertumor
cells (lung,suppressor,
osteoblastoma,proliferation
ALL, follicular
lymphoma, etc)
hsa-miR-193b-3p28153836many tissues/cells,arious cancertumor
sem*ncells (prostate,suppressor
breast, melanoma,
myeloma, non
small cell lung,
etc)follicular
lymphoma)
hsa-miR-193b-5p28163837many tissues/cells,arious cancertumor
sem*ncells (prostate,suppressor
breast, melanoma,
myeloma, non
small cell lung,
etc) follicular
lymphoma)
hsa-miR-194-3p28173838kidney, livervarious cancers
hsa-miR-194-5p28183839kidney, livervarious cancers
hsa-miR-195-3p28193840breast, pancreas
(islet)
hsa-miR-195-5p28203841breast, pancreas
(islet)
hsa-miR-196a-3p28213842pancreaticvarious canceroncogenic,
cells, endometrialcells (pancreatic,tumor
tissues,osteosarcoma,suppressor
mesenchymal stemendometrial,
cellsAML etc)
hsa-miR-196a-5p28223843pancreaticvarious canceroncogenic,
cells, endometrialcells (pancreatic,tumor
tissues,osteosarcoma,suppressor
mesenchymal stemendometrial,
cellsAML etc)
hsa-miR-196b-3p28233844endometrial tissuesglioblastomaapoptosis
hsa-miR-196b-5p28243845endometrial tissuesglioblastomaapoptosis
hsa-miR-197228253846acute
lymphoblastic
leukemia
hsa-miR-197328263847acute
lymphoblastic
leukemia
hsa-miR-197-3p28273848blood (myeloid),various cancers
other tissues/cells(thyroid tumor,
leukemia, etc)
hsa-miR-197-5p28283849blood (myeloid),various cancers
other tissues/cells(thyroid tumor,
leukemia, etc)
hsa-miR-197628293850acute
lymphoblastic
leukemia
hsa-miR-19828303851central nevous
system (CNS)
hsa-miR-199a-3p28313852liver, embryoid
body cells,
cardiomyocytes
hsa-miR-199a-5p28323853liver,
cardiomyocytes
hsa-miR-199b-3p28333854liver, osteoblastvarious cancersosteogenesis
hsa-miR-199b-5p28343855liver, osteoblastvarious cancersosteogenesis
hsa-miR-19a-3p28353856endothelial cellstumor
angiogenesis
hsa-miR-19a-5p28363857endothelial cellstumor
angiogenesis
hsa-miR-19b-1-5p28373858endothelial cellstumor
angiogenesis
hsa-miR-19b-2-5p28383859endothelial cellstumor
angiogenesis
hsa-miR-19b-3p28393860endothelial cellstumor
angiogenesis
hsa-miR-200a-3p28403861epithelial cells,various cancerstumor
many other tissues(breast, cervical,progression and
bladder, etc)metastasis
hsa-miR-200a-5p28413862epithelial cells,various cancerstumor
many other tissues(breast, cervical,progression and
bladder, etc)metastasis
hsa-miR-200b-3p28423863epithelial cells,tumor
many other tissuesprogression and
metastasis
hsa-miR-200b-5p28433864epithelial cells,tumor
many other tissuesprogression and
metastasis
hsa-miR-200c-3p28443865epithelial cells,tumor
many other tissues,progression and
embryonic stemmetastasis
cells
hsa-miR-200c-5p28453866epithelial cells,tumor
many other tissues,progression and
embryonic stemmetastasis
cells
hsa-miR-202-3p28463867bloodlymphomagenesis,
other cancers
hsa-miR-202-5p28473868bloodlymphomagenesis,
other cancers
hsa-miR-203a28483869skin (epithelium)psoriasis,
autoimmune
hsa-miR-203b-3p28493870skin specificpsoriasis,
(epithelium)autoimmune
hsa-miR-203b-5p28503871skin specificpsoriasis,
(epithelium)autoimmune
hsa-miR-204-3p28513872adipose, othervarious cancerstumor
tissues/cells, kidneymetastasis
hsa-miR-204-5p28523873adipose, othervarious cancerstumor
tissues/cells, kidneymetastasis
hsa-miR-205228533874
hsa-miR-205328543875
hsa-miR-205-3p28553876blood (plasma)various cancer
cells (breast,
glioma,
melanoma,
endometrial, etc)
hsa-miR-205428563877
hsa-miR-205-5p28573878blood (plasma)various cancer
cells (breast,
glioma,
melanoma,
endometrial, etc)
hsa-miR-20628583879muscle (cardiac andmyogenesis
skeletal)
hsa-miR-208a28593880heart (cardiomyocyte),cardiac defects
muscle
hsa-miR-208b28603881heart (cardiomyocyte),cardiac defects
muscle
hsa-miR-20a-3p28613882endothelial cells,
kidney, osteogenic
cells
hsa-miR-20a-5p28623883endothelial cells,
kidney, osteogenic
cells
hsa-miR-20b-3p28633884osteogenic cells
hsa-miR-20b-5p28643885osteogenic cells
hsa-miR-21028653886kidney, heart,RCC, B-cellangiogenesis
vascular endotheliallymphocytes
cells
hsa-miR-211028663887rectal cancer
hsa-miR-211328673888embryonic stem
cells
hsa-miR-211-3p28683889melanocytesmelanoma and
other cancers
hsa-miR-2114-3p28693890ovary, female
reproductuve tract
hsa-miR-2114-5p28703891ovary, female
reproductuve tract
hsa-miR-2115-3p28713892female reproductiveovarian cancer
tract
hsa-miR-2115-5p28723893female reproductiveovarian cancer
tract
hsa-miR-211-5p28733894melanocytesmelanoma and
other cancers
hsa-miR-2116-3p28743895live
cancer (hepatocytes)
and ovarian
cancer
hsa-miR-2116-5p28753896live
cancer (hepatocytes)
and ovarian
cancer
hsa-miR-211728763897ovarian cancer
hsa-miR-212-3p28773898brain (neuron),lymphoma
spleen
hsa-miR-212-5p28783899brain (neuron),lymphoma
spleen
hsa-miR-21-3p28793900glioblast, Bloodautoimmune,
(meyloid cells),heart diseases,
liver, vascularcancers
endothelial cells
hsa-miR-214-3p28803901immune cerlls,varioua cancersimmune
pancreas(melanoma,response
pancreatic,
ovarian)
hsa-miR-214-5p28813902immune cells,varioua cancersimmune
pancreas(melanoma,response
pancreatic,
ovarian)
hsa-miR-21528823903many tissues/cellsvarious cancerscell cycle
(renal, colon,arrest/p53
osteosarcoma)inducible
hsa-miR-21-5p28833904blood (myeloidautoimmune,
cells), liver,heart diseases,
endothelial cellscancers
hsa-miR-216a-3p28843905kidney, pancreas
hsa-miR-216a-5p28853906kidney, pancreas
hsa-miR-216b28863907cancerssenescence
hsa-miR-21728873908endothelial cellsvarious cancer
cells (pancreas,
kidney, breast)
hsa-miR-218-1-3p28883909endothelial cellsvarious cancer
cells (gastric
tumor, bladder,
cervical, etc)
hsa-miR-218-2-3p28893910various cancer
cells (gastric
tumor, bladder,
cervical, etc)
hsa-miR-218-5p28903911various cancer
cells (gastric
tumor, bladder,
cervical, etc)
hsa-miR-219-1-3p28913912brain,
oligodendrocytes
hsa-miR-219-2-3p28923913brain,
oligodendrocytes
hsa-miR-219-5p28933914brain,
oligodendrocytes
hsa-miR-221-3p28943915endothelial cells,leukemia andangiogenesis/vasculogenesis
immune cellsother cancers
hsa-miR-221-5p28953916endothelial cells,leukemia andangiogenesis/vasculogenesis
immune cellsother cancers
hsa-miR-222-3p28963917endothelial cellsvarious cancersangiogenesis
hsa-miR-222-5p28973918endothelial cellsvarious cancersangiogenesis
hsa-miR-223-3p28983919meyloid cellsleukemia
hsa-miR-223-5p28993920meyloid cellsleukemia
hsa-miR-22-3p29003921many tissues/cellsvarious cancerstumorigenesis
hsa-miR-224-3p29013922blood (plasma),cancers and
ovaryinflammation
hsa-miR-224-5p29023923blood (plasma),cancers and
ovaryinflammation
hsa-miR-22-5p29033924many tissues/cellsVarious cancerstumorigenesis
hsa-miR-227629043925breast cancer
hsa-miR-2277-3p29053926female reproductive
tract
hsa-miR-2277-5p29063927female reproductive
tract
hsa-miR-227829073928breast cancer
hsa-miR-2355-3p29083929embryonic stem
cells
hsa-miR-2355-5p29093930embryonic stem
cells
hsa-miR-239229103931identified in B-cells
hsa-miR-23a-3p29113932brain (astrocyte),Cancers
endothelial cells,
blood(erythroid)
hsa-miR-23a-5p29123933brain (astrocyte),cancers
endothelial cells,
blood (erythroid)
hsa-miR-23b-3p29133934blood, meyloidcancers (renal
cellscancer,
glioblastoma,
prostate, etc)
and autoimmune
hsa-miR-23b-5p29143935blood, meyloidcancers (glioblastoma,
cellsprostate, etc)
and autoimmune
hsa-miR-23c29153936cervical cancer
hsa-miR-24-1-5p29163937lung, meyloid cells
hsa-miR-24-2-5p29173938lung, meyloid cells
hsa-miR-24-3p29183939lung, meyloid cells
hsa-miR-2467-3p29193940breast cancer
hsa-miR-2467-5p29203941breast cancer
hsa-miR-25-3p29213942embryonic stem
cells, airway
smooth muscle
hsa-miR-25-5p29223943embryonic stem
cells, airway
smooth muscle
hsa-miR-2681-3p29233944breast cancer
hsa-miR-2681-5p29243945breast cancer
hsa-miR-2682-3p29253946
hsa-miR-2682-5p29263947
hsa-miR-26a-1-3p29273948embryonic stemCLL and othercell cycle and
cells, blood, othercancersdifferentiation
tissues
hsa-miR-26a-2-3p29283949blood, other tissuesCLL and othercell cycle and
cancersdifferentiation
hsa-miR-26a-5p29293950blood, other tissuesCLL and othercell cycle and
cancersdifferentiation
hsa-miR-26b-3p29303951hematopoietic cells
hsa-miR-26b-5p29313952hematopoietic cells
hsa-miR-27a-3p29323953meyloid cellsvarious cancer
cells
hsa-miR-27a-5p29333954meyloid cellsvarious cancer
cells
hsa-miR-27b-3p29343955meyloid cells,various cancerpro-angiogenic
vascular endothelialcells
cells
hsa-miR-27b-5p29353956meyloid cells,various cancerpro-angiogenic
vascular endothelialcells
cells
hsa-miR-28-3p29363957blood (immuneB/T cell
cells)lymphoma
hsa-miR-28-5p29373958blood (immuneB/T cell
cells)lymphoma
hsa-miR-286129383959osteoblastsbasal cell
carcinoma
hsa-miR-290929393960T-Lymphocytes
hsa-miR-296-3p29403961kidney, heart, lung,angiogenesis
entothelial cells
hsa-miR-2964a-3p29413962
hsa-miR-2964a-5p29423963
hsa-miR-296-5p29433964lung, liver,angiogenesis
endothelial cells
hsa-miR-29729443965oocyte and prostate
hsa-miR-29829453966breast cancer
hsa-miR-299-3p29463967myeloid
leukaemia,
hepatoma, breast
cancer
hsa-miR-299-5p29473968myeloid
leukaemia,
hepatoma, breast
cancer
hsa-miR-29a-3p29483969immuno systemCLL, othertumor
cancers,suppression,
neurodegenativeimmune
diseasemodulation
hsa-miR-29a-5p29493970immuno systemCLL, othertumor
cancers,suppression,
neurodegenativeimmune
diseasemodulation
hsa-miR-29b-1-5p29503971immuno systemCLL, othertumor
cancers,suppression,
neurodegenativeimmune
diseasemodulation
hsa-miR-29b-2-5p29513972immuno systemCLL, othertumor
cancerssuppression,
immune
modulation
hsa-miR-29b-3p29523973immuno systemCLL, othertumor
cancerssuppression,
immune
modulation
hsa-miR-29c-3p29533974immuno systemCLL, othertumor
cancerssuppression,
immune
modulation
hsa-miR-29c-5p29543975immuno systemCLL, othertumor
cancerssuppression,
immune
modulation
hsa-miR-30029553976osteoblastBladder cancer
hsa-miR-301a-3p29563977embryonic stem
cells
hsa-miR-301a-5p29573978embryonic stem
cells
hsa-miR-301b29583979esophageal
adenocarcinoma,
colonic cancer
hsa-miR-302a-3p29593980embryonic stemlipid
cells, lipidmetabolism
metabolism
hsa-miR-302a-5p29603981embryonic stemlipid
cells, lipidmetabolism
metabolism
hsa-miR-302b-3p29613982embryonic stem
cells
hsa-miR-302b-5p29623983embryonic stem
cells
hsa-miR-302c-3p29633984embryonic stem
cells
hsa-miR-302c-5p29643985embryonic stem
cells
hsa-miR-302d-3p29653986embryonic stem
cells
hsa-miR-302d-5p29663987embryonic stem
cells
hsa-miR-302e29673988embryoid body
cells
hsa-miR-302f29683989gastric cancer
hsa-miR-3064-3p29693990
hsa-miR-3064-5p29703991
hsa-miR-3065-3p29713992oligodendrocytesanti-virus
response
hsa-miR-3065-5p29723993oligodendrocytessolid tumors
hsa-miR-3074-3p29733994various
cancer (melanoma,
breast)
hsa-miR-3074-5p29743995various
cancer (melanoma,
breast)
hsa-miR-30a-3p29753996kidney, pancreaticvarious cancersautophagy
cells
hsa-miR-30a-5p29763997CNS (prefrontalglioma, colonautophagy
cortex), othercarcinoma
tissues
hsa-miR-30b-3p29773998kidney, adipose,
CNS (prefrontal
cortex)
hsa-miR-30b-5p29783999kidney, adipose,
CNS (prefrontal
cortex)
hsa-miR-30c-1-3p29794000kidney, adipose,
CNS (prefrontal
cortex)
hsa-miR-30c-2-3p29804001kidney, adipose,
CNS (prefrontal
cortex)
hsa-miR-30c-5p29814002kidney, adipose,
CNS (prefrontal
cortex)
hsa-miR-30d-3p29824003CNS (prefrontal
cortex
hsa-miR-30d-5p29834004CNS (prefrontal
cortex, embryoid
body cells
hsa-miR-30e-3p29844005myeloid cells, glia
cells
hsa-miR-30e-5p29854006myeloid cells, glia
cells
hsa-miR-311529864007various cancer
(melanoma,
breast tumor)
hsa-miR-311629874008discovered in the
melanoma
miRNAome
hsa-miR-3117-3p29884009discovered in the
melanoma
miRNAome
hsa-miR-3117-5p29894010discovered in the
melanoma
miRNAome
hsa-miR-311829904011discovered in the
melanoma
miRNAome
hsa-miR-311929914012discovered in the
melanoma
miRNAome
hsa-miR-3120-3p29924013discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3120-5p29934014discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3121-3p29944015discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3121-5p29954016discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-312229964017discovered in the
melanoma
miRNAome
hsa-miR-312329974018discovered in the
melanoma
miRNAome
hsa-miR-3124-3p29984019discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3124-5p29994020discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-312530004021discovered in the
melanoma
miRNAome
hsa-miR-3126-3p30014022discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3126-5p30024023discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3127-3p30034024discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3127-5p30044025discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-312830054026discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3129-3p30064027discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3129-5p30074028discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3130-3p30084029discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3130-5p30094030discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-313130104031discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-313230114032discovered in the
melanoma
miRNAome
hsa-miR-313330124033discovered in the
melanoma
miRNAome
hsa-miR-313430134034discovered in the
melanoma
miRNAome
hsa-miR-3135a30144035discovered in the
melanoma
miRNAome
hsa-miR-3135b30154036discovered in B
cells
hsa-miR-3136-3p30164037discovered in thelymphoblastic
melanomaleukaemia and
miRNAomebreast tumor
hsa-miR-3136-5p30174038discovered in thelymphoblastic
melanomaleukaemia and
miRNAomebreast tumor
hsa-miR-313730184039discovered in the
melanoma
miRNAome
hsa-miR-313830194040discovered in the
melanoma
miRNAome, ovary
hsa-miR-313930204041discovered in the
melanoma
miRNAome
hsa-miR-31-3p30214042
hsa-miR-3140-3p30224043discovered in thelymphoblastic
melanomaleukaemia and
miRNAome, ovarybreast tumor
hsa-miR-3140-5p30234044discovered in thelymphoblastic
melanomaleukaemia and
miRNAome, ovarybreast tumor
hsa-miR-314130244045discovered in the
melanoma
miRNAome
hsa-miR-314230254046discovered in the
melanoma
miRNAome;
immune cells
hsa-miR-314330264047discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3144-3p30274048discovered in the
melanoma
miRNAome, ovary
hsa-miR-3144-5p30284049discovered in the
melanoma
miRNAome, ovary
hsa-miR-3145-3p30294050discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3145-5p30304051discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-314630314052discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-314730324053discovered in the
melanoma
miRNAome
hsa-miR-314830334054discovered in the
melanoma
miRNAome
hsa-miR-314930344055discovered in the
melanoma
miRNAome, ovary
hsa-miR-3150a-3p30354056discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3150a-5p30364057discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3150b-3p30374058discovered in thebreast tumor and
melanomalymphoblastic
miRNAomeleukaemia
hsa-miR-3150b-5p30384059discovered in thebreast tumor and
melanomalymphoblastic
miRNAomeleukaemia
hsa-miR-315130394060discovered in thelymphoblastic
melanomaleukaemia
miRNAome
hsa-miR-3152-3p30404061discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3152-5p30414062discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-315330424063discovered in the
melanoma
miRNAome
hsa-miR-315430434064discovered in thelymphoblastic
melanomaleukaemia
miRNAome
hsa-miR-3155a30444065discovered in the
melanoma
miRNAome
hsa-miR-3155b30454066discovered in B
cells
hsa-miR-3156-3p30464067discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3156-5p30474068discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3157-3p30484069discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3157-5p30494070discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3158-3p30504071discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3158-5p30514072discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-315930524073discovered in the
melanoma
miRNAome
hsa-miR-31-5p30534074various cancer
cells (breast, lung,
prostate)
hsa-miR-3160-3p30544075discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3160-5p30554076discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-316130564077discovered in the
melanoma
miRNAome
hsa-miR-3162-3p30574078discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3162-5p30584079discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-316330594080discovered in the
melanoma
miRNAome
hsa-miR-316430604081discovered in the
melanoma
miRNAome
hsa-miR-316530614082discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-316630624083discovered in the
melanoma
miRNAome
hsa-miR-316730634084discovered in the
melanoma
miRNAome, ovary
hsa-miR-316830644085discovered in the
melanoma
miRNAome
hsa-miR-316930654086discovered in the
melanoma
miRNAome
hsa-miR-317030664087discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-317130674088discovered in the
melanoma
miRNAome, ovary
hsa-miR-3173-3p30684089discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3173-5p30694090discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-317430704091discovered in the
melanoma
miRNAome
hsa-miR-317530714092discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-317630724093discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3177-3p30734094discovered in thebreast tumor and
melanomalymphoblastic
miRNAomeleukaemia
hsa-miR-3177-5p30744095discovered in thebreast tumor and
melanomalymphoblastic
miRNAomeleukaemia
hsa-miR-317830754096discovered in the
melanoma
miRNAome
hsa-miR-317930764097discovered in the
melanoma
miRNAome
hsa-miR-318030774098discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3180-3p30784099discovered in breast
tunor
hsa-miR-3180-5p30794100discovered in breast
tumor
hsa-miR-318130804101discovered in the
melanoma
miRNAome
hsa-miR-318230814102discovered in the
melanoma
miRNAome
hsa-miR-318330824103discovered in the
melanoma
miRNAome
hsa-miR-3184-3p30834104discovered in the
melanoma
miRNAome
hsa-miR-3184-5p30844105discovered in the
melanoma
miRNAome
hsa-miR-318530854106discovered in the
melanoma
miRNAome
hsa-miR-3186-3p30864107discovered in the
melanoma
miRNAome, ovary
hsa-miR-3186-5p30874108discovered in the
melanoma
miRNAome, ovary
hsa-miR-3187-3p30884109discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3187-5p30894110discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-318830904111discovered in the
melanoma
miRNAome
hsa-miR-3189-3p30914112discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3189-5p30924113discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3190-3p30934114discovered in thelymphoblastic
melanomaleukaemia
miRNAome
hsa-miR-3190-5p30944115discovered in thelymphoblastic
melanomaleukaemia
miRNAome
hsa-miR-3191-3p30954116discovered in the
melanoma
miRNAome
hsa-miR-3191-5p30964117discovered in the
melanoma
miRNAome
hsa-miR-319230974118discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-319330984119discovered in the
melanoma
miRNAome
hsa-miR-3194-3p30994120discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-3194-5p31004121discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-319531014122discovered in the
melanoma
miRNAome
hsa-miR-319631024123basal cell
carcinoma
hsa-miR-319731034124discovered in the
melanoma
miRNAome
hsa-miR-319831044125discovered in thebreast tumor
melanoma
miRNAome
hsa-miR-319931054126discovered in the
melanoma
miRNAome
hsa-miR-3200-3p31064127discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-3200-5p31074128discovered in thebreast tumor
melanoma
miRNAome, ovary
hsa-miR-320131084129discovered in the
melanoma
miRNAome,
hsa-miR-320231094130discovered in the
melanoma
miRNAome, epithelial
cell BEAS2B
hsa-miR-320a31104131blood,colon cancer
heart (myocardiac)cells, heart
disease
hsa-miR-320b31114132central nevous
system
hsa-miR-320c31124133chondrocytecartilage
metabolism
hsa-miR-320d31134134cancer stem cells
hsa-miR-320e31144135neural cells
hsa-miR-323a-3p31154136neuronsmyeloid
leukaemia,
mudulla thyroid
carcinoma
hsa-miR-323a-5p31164137neuronsmyeloid
leukaemia,
mudulla thyroid
carcinoma
hsa-miR-323b-3p31174138myeloid
leukaemia
hsa-miR-323b-5p31184139myeloid
leukaemia
hsa-miR-32-3p31194140blood, gliavarious cancers
(lung, kidney,
prostate, etc),
virus infection
hsa-miR-324-3p31204141kidney
hsa-miR-324-5p31214142neuronstumor cells
hsa-miR-32531224143neurons, placenta
hsa-miR-32-5p31234144blood, gliavarious cancers
(lung, kidney,
prostate, etc),
virus infection
hsa-miR-32631244145neuronstumor cells
hsa-miR-32831254146neuron, bloodtumor cells
hsa-miR-32931264147brain and platele
hsa-miR-330-3p31274148various cancers
(prostate,
glioblastoma,
colorectal)
hsa-miR-330-5p31284149various cancers
(prostate,
glioblastoma,
colorectal)
hsa-miR-331-3p31294150gastric cancer
hsa-miR-331-5p31304151lymphocytes
hsa-miR-335-3p31314152kidney, breastRCC, multiple
myeloma
hsa-miR-335-5p31324153kidney, breastRCC, multiple
myeloma
hsa-miR-337-3p31334154lunggastric cancer
hsa-miR-337-5p31344155lung
hsa-miR-338-3p31354156epithelial cells,gastric, rectal
oligodendrocytescancer cells,
osteosarcoma
hsa-miR-338-5p31364157oligodendrocytesgastric cancer
hsa-miR-339-3p31374158immune cell
hsa-miR-339-5p31384159immune cell
hsa-miR-33a-3p31394160pancreatic islet,lipid
lipid metabolismmetabolism
hsa-miR-33a-5p31404161pancreatic islet,lipid
lipid metabolismmetabolism
hsa-miR-33b-3p31414162lipid metabolismlipid
metabolism
hsa-miR-33b-5p31424163lipid metabolismlipid
metabolism
hsa-miR-340-3p31434164various cancers
hsa-miR-340-5p31444165embryoid body
cells
hsa-miR-342-3p31454166brain, circulatingmultiple
plasmamyeloma, other
cancers
hsa-miR-342-5p31464167circulating plasmamultiple
myeloma, other
cancers
hsa-miR-345-3p31474168hematopoietic cellsfollicular
lymphoma, other
cancers
hsa-miR-345-5p31484169hematopoietic cellsfollicular
lymphoma, other
cancers
hsa-miR-34631494170immume cellscancers and
autoimmune
hsa-miR-34a-3p31504171breast, meyloidgastric cancer,tumor
cells, ciliatedCLL, othersuppressor, p53
epithelial cellsinducible
hsa-miR-34a-5p31514172breast, meyloidgastric cancer,tumor
cells, ciliatedCLL, othersuppressor, p53
epithelial cellsinducible
hsa-miR-34b-3p31524173ciliated epithelialvarious cancerstumor
cellssuppressor, p53
inducible
hsa-miR-34b-5p31534174ciliated epithelialvarious cancerstumor
cellssuppressor, p53
inducible
hsa-miR-34c-3p31544175ciliated epithelialvarious cancerstumor
cells, placentasuppressor, p53
inducible
hsa-miR-34c-5p31554176ciliated epithelialvarious cancerstumor
cells, placentasuppressor, p53
inducible
hsa-miR-3529-3p31564177discovered in breast
tumor
hsa-miR-3529-5p31574178discovered in breast
tumor
hsa-miR-3591-3p31584179discovered in breast
tumor
hsa-miR-3591-5p31594180discovered in breast
tumor
hsa-miR-3605-3p31604181discovered in
reprodcutive tracts
hsa-miR-3605-5p31614182discovered in
reprodcutive tracts
hsa-miR-3606-3p31624183discovered in
cervical tumors
hsa-miR-3606-5p31634184discovered in
cervical tumors
hsa-miR-3607-3p31644185discovered in
cervical tumors
hsa-miR-3607-5p31654186discovered in
cervical tumors
hsa-miR-360931664187discovered in
cervical tumors
hsa-miR-361031674188discovered in
cervical tumors
hsa-miR-361131684189discovered in
cervical tumors
hsa-miR-361231694190discovered in
cervical tumors
hsa-miR-3613-3p31704191discovered in
cervical tumors
hsa-miR-3613-5p31714192discovered in
cervical tumors
hsa-miR-361-3p31724193blood, endothelial
cells
hsa-miR-3614-3p31734194discovered in
cervical and breast
tumors
hsa-miR-3614-5p31744195discovered in
cervical and breast
tumors
hsa-miR-361531754196discovered in
cervical tumors
hsa-miR-361-5p31764197endothelial cells
hsa-miR-3616-3p31774198discovered in
cervical tumors
hsa-miR-3616-5p31784199discovered in
cervical tumors
hsa-miR-3617-3p31794200discovered in
cervical tumors and
psoriasis
hsa-miR-3617-5p31804201discovered in
cervical tumors and
psoriasis
hsa-miR-361831814202discovered in
cervical tumors
hsa-miR-3619-3p31824203discovered in breast
tumors
hsa-miR-3619-5p31834204discovered in breast
tumors
hsa-miR-3620-3p31844205discovered in
cervical tumors
hsa-miR-3620-5p31854206discovered in
cervical tumors
hsa-miR-362131864207discovered in
cervical tumors
hsa-miR-3622a-3p31874208discovered in breast
tumors
hsa-miR-3622a-5p31884209discovered in breast
tumors
hsa-miR-3622b-3p31894210discovered in
cervical tumors
hsa-miR-3622b-5p31904211discovered in
cervical tumors
hsa-miR-362-3p31914212melanoma
hsa-miR-362-5p31924213melanoma
hsa-miR-363-3p31934214kidney stem cell,
blood cells
hsa-miR-363-5p31944215kidney stem cell,
blood cells
hsa-miR-364631954216discovered in solid
tumor
hsa-miR-364831964217discovered in solid
tumor
hsa-miR-364931974218discovered in solid
tumor
hsa-miR-365031984219discovered in solid
tumor
hsa-miR-365131994220discovered in solid
tumor
hsa-miR-365232004221discovered in solid
tumor
hsa-miR-365332014222discovered in solid
tumor
hsa-miR-365432024223discovered in solid
tumor
hsa-miR-365532034224discovered in solid
tumor
hsa-miR-365632044225discovered in solid
tumor
hsa-miR-365732054226discovered in solid
tumor
hsa-miR-365832064227discovered in solid
tumor
hsa-miR-365932074228discovered in breast
tumors
hsa-miR-365a-3p32084229various cancerapoptosis
cells (Immune
cells, lung, colon,
endometriotic)
hsa-miR-365a-5p32094230various cancerapoptosis
cells (Immune
cells, lung, colon,
endometriotic))
hsa-miR-365b-3p32104231various cancersapoptosis
(retinoblastoma, colon,
endometriotic)
hsa-miR-365b-5p32114232various cancersapoptosis
(colon,
endometriotic)
hsa-miR-366032124233discovered in breast
tumors
hsa-miR-366132134234discovered in breast
tumors
hsa-miR-366232144235
hsa-miR-3663-3p32154236
hsa-miR-3663-5p32164237
hsa-miR-3664-3p32174238discovered in breast
tumors
hsa-miR-3664-5p32184239discovered in breast
tumors
hsa-miR-366532194240brain
hsa-miR-366632204241brain
hsa-miR-3667-3p32214242discovered in
peripheral blood
hsa-miR-3667-5p32224243discovered in
peripheral blood
hsa-miR-366832234244discovered in
peripheral blood
hsa-miR-366932244245discovered in
peripheral blood
hsa-miR-367032254246discovered in
peripheral blood
hsa-miR-367132264247discovered in
peripheral blood
hsa-miR-367232274248discovered in
peripheral blood
hsa-miR-367332284249discovered in
peripheral blood
hsa-miR-367-3p32294250embryonic stemreprogramming
cells
hsa-miR-367432304251discovered in
peripheral blood
hsa-miR-3675-3p32314252discovered in
peripheral blood
hsa-miR-3675-5p32324253discovered in
peripheral blood
hsa-miR-367-5p32334254embryonic stemreprogramming
cells
hsa-miR-3676-3p32344255discovered in
peripheral blood
hsa-miR-3676-5p32354256discovered in
peripheral blood
hsa-miR-3677-3p32364257discovered in
peripheral blood
hsa-miR-3677-5p32374258discovered in
peripheral blood
hsa-miR-3678-3p32384259discovered in
peripheral blood
hsa-miR-3678-5p32394260discovered in
peripheral blood
hsa-miR-3679-3p32404261discovered in
peripheral blood
hsa-miR-3679-5p32414262discovered in
peripheral blood
hsa-miR-3680-3p32424263discovered in
peripheral blood
hsa-miR-3680-5p32434264discovered in
peripheral blood
hsa-miR-3681-3p32444265discovered in
peripheral blood
hsa-miR-3681-5p32454266discovered in
peripheral blood
hsa-miR-3682-3p32464267discovered in
peripheral blood
hsa-miR-3682-5p32474268discovered in
peripheral blood
hsa-miR-368332484269discovered in
peripheral blood
hsa-miR-368432494270discovered in
peripheral blood
hsa-miR-368532504271discovered in
peripheral blood
hsa-miR-368632514272discovered in
peripheral blood
hsa-miR-368732524273discovered in
peripheral blood
hsa-miR-3688-3p32534274discovered in breast
tumor
hsa-miR-3688-5p32544275discovered in breast
tumor
hsa-miR-3689a-3p32554276discovered in
female
reproductuve tract
hsa-miR-3689a-5p32564277discovered in
female
reproductuve tract
and peripheral
blood
hsa-miR-3689b-3p32574278discovered in
female
reproductuve tract
and peripheral
blood
hsa-miR-3689b-5p32584279discovered in
female
reproductuve tract
hsa-miR-3689c32594280discovered in B
cells
hsa-miR-3689d32604281discovered in B
cells
hsa-miR-3689e32614282discovered in B
cells
hsa-miR-3689f32624283discovered in B
cells
hsa-miR-369032634284discovered in
peripheral blood
hsa-miR-3691-3p32644285discovered in
peripheral blood
hsa-miR-3691-5p32654286discovered in
peripheral blood
hsa-miR-3692-3p32664287discovered in
peripheral blood
hsa-miR-3692-5p32674288discovered in
peripheral blood
hsa-miR-369-3p32684289stem cellsreprogramming
hsa-miR-369-5p32694290stem cellsreprogramming
hsa-miR-37032704291acute meyloidtumor
leukaemia andsuppressor, lipid
other cancersmetabolism
hsa-miR-371332714292discovered in
neuroblastoma
hsa-miR-371432724293discovered in
neuroblastoma
hsa-miR-371a-3p32734294serum
hsa-miR-371a-5p32744295serum
hsa-miR-371b-3p32754296serum
hsa-miR-371b-5p32764297serum
hsa-miR-37232774298hematopoietic cells,
lung, placental
(blood)
hsa-miR-373-3p32784299breast cancer
hsa-miR-373-5p32794300breast cancer
hsa-miR-374a-3p32804301muscle (myoblasts)breast and lungmyogenic
cancerdifferentiation
hsa-miR-374a-5p32814302muscle (myoblasts)breast and lungmyogenic
cancerdifferentiation
hsa-miR-374b-3p32824303muscle (myoblasts)myogenic
differentiation
hsa-miR-374b-5p32834304muscle (myoblasts)myogenic
differentiation
hsa-miR-374c-3p32844305muscle (myoblasts)myogenic
differentiation
hsa-miR-374c-5p32854306muscle (myoblasts)myogenic
differentiation
hsa-miR-37532864307pancreas (islet)
hsa-miR-376a-2-5p32874308regulatory miRs for
hematopoietic cells
(erythroid, platelet,
lympho)
hsa-miR-376a-3p32884309regulatory miRs for
hematopoietic cells
(erythroid, platelet,
lympho)
hsa-miR-376a-5p32894310regulatory miRs for
hematopoietic cells
(erythroid, platelet,
lympho)
hsa-miR-376b-3p32904311bloodvarious cancerautophagy
cells
hsa-miR-376b-5p32914312bloodvarious cancerautophagy
cells
hsa-miR-376c-3p32924313trophoblastvarious cancercell proliferatio
cells
hsa-miR-376c-5p32934314trophoblastvarious cancercell proliferatio
cells
hsa-miR-377-3p32944315hematopoietic cells
hsa-miR-377-5p32954316hematopoietic cells
hsa-miR-378a-3p32964317ovary, lipid
metabolism
hsa-miR-378a-5p32974318ovary,
placenta/trophoblast,
lipid metabolism
hsa-miR-378b32984319lipid metabolism
hsa-miR-378c32994320lipid metabolism
hsa-miR-378d33004321lipid metabolism
hsa-miR-378e33014322lipid metabolism
hsa-miR-378f33024323lipid metabolism
hsa-miR-378g33034324lipid metabolism
hsa-miR-378h33044325lipid metabolism
hsa-miR-378i33054326lipid metabolism
hsa-miR-378j33064327lipid metabolism
hsa-miR-379-3p33074328various cancers
(breast,
hepatocytes,
colon)
hsa-miR-379-5p33084329various cancers
(breast,
hepatocytes,
colon)
hsa-miR-380-3p33094330brainneuroblastoma
hsa-miR-380-5p33104331brain, embryonicneuroblastoma
stem cells
hsa-miR-381-3p33114332chondrogenesis,
lung, brain
hsa-miR-381-5p33124333chondrogenesis,
lung, brain
hsa-miR-382-3p33134334renal epithelial cells
hsa-miR-382-5p33144335renal epithelial cells
hsa-miR-38333154336testes, brain
(medulla)
hsa-miR-38433164337epithelial cells
hsa-miR-390733174338discovered in
female reproductive
tract
hsa-miR-390833184339discovered in
female reproductive
tract
hsa-miR-390933194340discovered in
female reproductive
tract
hsa-miR-391033204341discovered in
female reproductive
tract
hsa-miR-391133214342discovered in breast
tumor and female
reproductive tract
hsa-miR-391233224343discovered in
female reproductive
tract
hsa-miR-3913-3p33234344discovered in breast
tumor and female
reproductive tract
hsa-miR-3913-5p33244345discovered in breast
tumor and female
reproductive tract
hsa-miR-391433254346discovered in breast
tumor and female
reproductive tract
hsa-miR-391533264347discovered in
female reproductive
tract
hsa-miR-391633274348discovered in
female reproductive
tract
hsa-miR-391733284349discovered in
female reproductive
tract
hsa-miR-391833294350discovered in
female reproductive
tract
hsa-miR-391933304351discovered in
female reproductive
tract
hsa-miR-392033314352discovered in
female reproductive
tract
hsa-miR-392133324353discovered in
female reproductive
tract
hsa-miR-3922-3p33334354discovered in breast
tumor and female
reproductive tract
hsa-miR-3922-5p33344355discovered in breast
tumor and female
reproductive tract
hsa-miR-392333354356discovered in
female reproductive
tract
hsa-miR-392433364357discovered in
female reproductive
tract
hsa-miR-3925-3p33374358discovered in breast
tumor and female
reproductive tract
hsa-miR-3925-5p33384359discovered in breast
tumor and female
reproductive tract
hsa-miR-392633394360discovered in
female reproductive
tract
hsa-miR-3927-3p33404361discovered in
female reproductive
tract and psoriasis
hsa-miR-3927-5p33414362discovered in
female reproductive
tract and psoriasis
hsa-miR-392833424363discovered in
female reproductive
tract
hsa-miR-392933434364discovered in
female reproductive
tract
hsa-miR-3934-3p33444365discovered in
abnormal skin
(psoriasis)
hsa-miR-3934-5p33454366discovered in
abnormal skin
(psoriasis)
hsa-miR-393533464367
hsa-miR-393633474368discovered in breast
tumor and
lymphoblastic
leukaemia
hsa-miR-393733484369
hsa-miR-393833494370
hsa-miR-393933504371
hsa-miR-3940-3p33514372discovered in breast
tumor
hsa-miR-3940-5p33524373discovered in breast
tumor
hsa-miR-394133534374
hsa-miR-3942-3p33544375discovered in breast
tumor and
lymphoblastic
leukaemia
hsa-miR-3942-5p33554376discovered in breast
tumor and
lymphoblastic
leukaemia
hsa-miR-394333564377
hsa-miR-3944-3p33574378discovered in breast
tumor
hsa-miR-3944-5p33584379discovered in breast
tumor
hsa-miR-394533594380
hsa-miR-396033604381osteoblast
hsa-miR-397233614382discovered in Acute
Myeloid Leukaemia
hsa-miR-397333624383discovered in Acute
Myeloid Leukaemia
hsa-miR-397433634384discovered in Acute
Myeloid Leukaemia
hsa-miR-397533644385discovered in Acute
Myeloid Leukaemia
hsa-miR-397633654386discovered in Acute
Myeloid Leukaemia
hsa-miR-397733664387discovered in Acute
Myeloid Leukaemia
hsa-miR-397833674388discovered in Acute
Myeloid Leukaemia
hsa-miR-409-3p33684389gastric cancer
hsa-miR-409-5p33694390gastric cancer
hsa-miR-41033704391brainglioma
hsa-miR-411-3p33714392Glioblastoma
others
hsa-miR-411-5p33724393Glioblastoma
others
hsa-miR-41233734394upregulated in
lung cancer
hsa-miR-42133744395endothelial cellsgastric cancer,
HCC
hsa-miR-422a33754396circulating
microRNA (in
plasma)
hsa-miR-423-3p33764397embryonic stem
cells
hsa-miR-423-5p33774398heart, embryonic
stem cells
hsa-miR-424-3p33784399endothelial cellsvariouspro-angiogenic
cancers (e.g B-
lieage ALL),
cardiac diseases
hsa-miR-424-5p33794400endothelial cellsvariouspro-angiogenic
cancers (e.g B-
lieage ALL),
cardiac diseases
hsa-miR-425133804401discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425233814402discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425333824403discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425-3p33834404brainovarian cancer,
brain tumor
hsa-miR-425433844405discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425533854406discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425-5p33864407brainB-lieage ALL,
brain tumor
hsa-miR-425633874408discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425733884409discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425833894410discovered in
embryonic stem
cells and neural
precusors
hsa-miR-425933904411discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426033914412discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426133924413discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426233934414discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426333944415discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426433954416discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426533964417discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426633974418discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426733984419discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426833994420discovered in
embryonic stem
cells and neural
precusors
hsa-miR-426934004421discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427034014422discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427134024423discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427234034424discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427334044425
hsa-miR-427434054426discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427534064427discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427634074428discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427734084429discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427834094430discovered in
embryonic stem
cells and neural
precusors
hsa-miR-427934104431discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428034114432discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428134124433discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428234134434discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428334144435discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428434154436discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428534164437discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428634174438discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428734184439discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428834194440discovered in
embryonic stem
cells and neural
precusors
hsa-miR-428934204441discovered in
embryonic stem
cells and neural
precusors
hsa-miR-42934214442Epithelial cellsvarious cancers
(colorectal,
endometrial,
gastric, ovarian
etc)
hsa-miR-429034224443discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429134234444discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429234244445discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429334254446discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429434264447discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429534274448discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429634284449discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429734294450discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429834304451discovered in
embryonic stem
cells and neural
precusors
hsa-miR-429934314452discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430034324453discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430134334454discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430234344455discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430334354456discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430434364457discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430534374458discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430634384459discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430734394460discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430834404461discovered in
embryonic stem
cells and neural
precusors
hsa-miR-430934414462discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431034424463discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431134434464discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431234444465discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431334454466discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431-3p34464467Cancers
(follicular
lymphoma)
hsa-miR-431434474468discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431534484469discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431-5p34494470Cancers
(follicular
lymphoma)
hsa-miR-431634504471discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431734514472discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431834524473discovered in
embryonic stem
cells and neural
precusors
hsa-miR-431934534474discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432034544475discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432134554476discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432234564477discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432334574478discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432-3p34584479myoblastmyogenic
differentiation
hsa-miR-432434594480discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432534604481discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432-5p34614482myoblastmyogenic
differentiation
hsa-miR-432634624483discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432734634484discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432834644485discovered in
embryonic stem
cells and neural
precusors
hsa-miR-432934654486discovered in
embryonic stem
cells and neural
precusors
hsa-miR-43334664487various diseases
(cancer,
Parkinson's,
Chondrodysplasia)
hsa-miR-433034674488discovered in
embryonic stem
cells and neural
precusors
hsa-miR-441734684489discovered in B
cells
hsa-miR-441834694490discovered in B
cells
hsa-miR-4419a34704491discovered in B
cells
hsa-miR-4419b34714492discovered in B
cells
hsa-miR-442034724493discovered in B
cells
hsa-miR-442134734494discovered in B
cells
hsa-miR-442234744495discovered in breast
tumor and B cells
hsa-miR-4423-3p34754496discovered in breast
tumor, B cells and
skin(psoriasis)
hsa-miR-4423-5p34764497discovered in breast
tumor B cells and
skin(psoriasis)
hsa-miR-442434774498discovered in B
cells
hsa-miR-442534784499discovered in B
cells
hsa-miR-442634794500discovered in B
cells
hsa-miR-442734804501discovered in B
cells
hsa-miR-442834814502discovered in B
cells
hsa-miR-442934824503discovered in B
cells
hsa-miR-443034834504discovered in B
cells
hsa-miR-443134844505discovered in B
cells
hsa-miR-443234854506discovered in B
cells
hsa-miR-4433-3p34864507discovered in B
cells
hsa-miR-4433-5p34874508discovered in B
cells
hsa-miR-443434884509discovered in B
cells
hsa-miR-443534894510discovered in B
cells
hsa-miR-4436a34904511discovered in breast
tumor and B cells
hsa-miR-4436b-3p34914512discovered in breast
tumor
hsa-miR-4436b-5p34924513discovered in breast
tumor
hsa-miR-443734934514discovered in B
cells
hsa-miR-443834944515discovered in B
cells
hsa-miR-443934954516discovered in B
cells
hsa-miR-444034964517discovered in B
cells
hsa-miR-444134974518discovered in B
cells
hsa-miR-444234984519discovered in B
cells
hsa-miR-444334994520discovered in B
cells
hsa-miR-444435004521discovered in B
cells
hsa-miR-4445-3p35014522discovered in B
cells
hsa-miR-4445-5p35024523discovered in B
cells
hsa-miR-4446-3p35034524discovered in breast
tumor and B cells
hsa-miR-4446-5p35044525discovered in breast
tumor and B cells
hsa-miR-444735054526discovered in B
cells
hsa-miR-444835064527discovered in B
cells
hsa-miR-444935074528discovered in B
cells
hsa-miR-445035084529discovered in B
cells
hsa-miR-445135094530discovered in B
cells
hsa-miR-445235104531discovered in B
cells
hsa-miR-445335114532discovered in B
cells
hsa-miR-445435124533discovered in B
cells
hsa-miR-445535134534discovered in B
cells
hsa-miR-445635144535discovered in B
cells
hsa-miR-445735154536discovered in B
cells
hsa-miR-445835164537discovered in B
cells
hsa-miR-445935174538discovered in B
cells
hsa-miR-446035184539discovered in B
cells
hsa-miR-446135194540discovered in B
cells
hsa-miR-446235204541discovered in B
cells
hsa-miR-446335214542discovered in B
cells
hsa-miR-446435224543discovered in B
cells
hsa-miR-446535234544discovered in B
cells
hsa-miR-446635244545discovered in B
cells
hsa-miR-446735254546discovered in breast
tumor and B cells
hsa-miR-446835264547discovered in B
cells
hsa-miR-446935274548discovered in breast
tumor and B cells
hsa-miR-447035284549discovered in B
cells
hsa-miR-447145505571discovered in breast
tumor and B cells
hsa-miR-447245515572discovered in B
cells
hsa-miR-447345525573discovered in B
cells
hsa-miR-4474-3p45535574discovered in breast
tumor,
lymphoblastic
leukaemia and B
cells
hsa-miR-4474-5p45545575discovered in breast
tumor,
lymphoblastic
leukaemia and B
cells
hsa-miR-447545555576discovered in B
cells
hsa-miR-447645565577discovered in B
cells
hsa-miR-4477a45575578discovered in B
cells
hsa-miR-4477b45585579discovered in B
cells
hsa-miR-447845595580discovered in B
cells
hsa-miR-447945605581discovered in B
cells
hsa-miR-44845615582liver (hepatocytes)HCC
hsa-miR-448045625583discovered in B
cells
hsa-miR-448145635584discovered in B
cells
hsa-miR-4482-3p45645585discovered in B
cells
hsa-miR-4482-5p45655586discovered in B
cells
hsa-miR-448345665587discovered in B
cells
hsa-miR-448445675588discovered in B
cells
hsa-miR-448545685589discovered in B
cells
hsa-miR-448645695590discovered in B
cells
hsa-miR-448745705591discovered in B
cells
hsa-miR-448845715592discovered in B
cells
hsa-miR-448945725593discovered in breast
tumor and B cells
hsa-miR-449045735594discovered in B
cells
hsa-miR-449145745595discovered in B
cells
hsa-miR-449245755596discovered in B
cells
hsa-miR-449345765597discovered in B
cells
hsa-miR-449445775598discovered in B
cells
hsa-miR-449545785599discovered in B
cells
hsa-miR-449645795600discovered in B
cells
hsa-miR-449745805601discovered in B
cells
hsa-miR-449845815602discovered in B
cells
hsa-miR-449945825603discovered in B
cells
hsa-miR-449a45835604chondrocytes, ciliatedlung, colonic,cell cycle
epithelial cellsovarian cancerprogression and
proliferation
hsa-miR-449b-3p45845605ciliated epithelialvarious cancercell cycle
cells, other tissuescellsprogression and
proliferation
hsa-miR-449b-5p45855606ciliated epithelialvarious cancercell cycle
cells, other tissuescellsprogression and
proliferation
hsa-miR-449c-3p45865607epithelial ovarian
cancer cells
hsa-miR-449c-5p45875608epithelial ovarian
cancer cells
hsa-miR-450045885609discovered in B
cells
hsa-miR-450145895610discovered in B
cells
hsa-miR-450245905611discovered in B
cells
hsa-miR-450345915612discovered in B
cells
hsa-miR-450445925613discovered in B
cells
hsa-miR-450545935614discovered in B
cells
hsa-miR-450645945615discovered in B
cells
hsa-miR-450745955616discovered in B
cells
hsa-miR-450845965617discovered in B
cells
hsa-miR-450945975618discovered in B
cells
hsa-miR-450a-3p45985619
hsa-miR-450a-5p45995620
hsa-miR-450b-3p46005621
hsa-miR-450b-5p46015622
hsa-miR-451046025623discovered in B
cells
hsa-miR-451146035624discovered in B
cells
hsa-miR-451246045625discovered in B
cells
hsa-miR-451346055626discovered in B
cells
hsa-miR-451446065627discovered in B
cells
hsa-miR-451546075628discovered in B
cells
hsa-miR-451646085629discovered in B
cells
hsa-miR-451746095630discovered in B
cells
hsa-miR-451846105631discovered in B
cells
hsa-miR-451946115632discovered in B
cells
hsa-miR-451a46125633heart, central
nevous system,
epithelial cells
hsa-miR-451b46135634heart, central
nevous system,
epithelial cells
hsa-miR-4520a-3p46145635discovered in breast
tumor and B cells,
skin (psoriasis)
hsa-miR-4520a-5p46155636discovered in breast
tumor and B cells,
skin (psoriasis)
hsa-miR-4520b-3p46165637discovered in breast
tumor
hsa-miR-4520b-5p46175638discovered in breast
tumor
hsa-miR-452146185639discovered in B
cells
hsa-miR-452246195640discovered in B
cells
hsa-miR-452346205641discovered in B
cells
hsa-miR-452-3p46215642myoblastbladder cancer
and others
hsa-miR-4524a-3p46225643discovered in breast
tumor and B cells,
skin (psoriasis)
hsa-miR-4524a-5p46235644discovered in breast
tumor and B cells,
skin (psoriasis)
hsa-miR-4524b-3p46245645discovered in breast
tumor and B cells,
skin (psoriasis)
hsa-miR-4524b-5p46255646discovered in breast
tumor and B cells,
skin (psoriasis)
hsa-miR-452546265647discovered in B
cells
hsa-miR-452-5p46275648myoblastbladder cancer
and others
hsa-miR-452646285649discovered in breast
tumor and B cells
hsa-miR-452746295650discovered in B
cells
hsa-miR-452846305651discovered in B
cells
hsa-miR-4529-3p46315652discovered in breast
tumor and B cells
hsa-miR-4529-5p46325653discovered in breast
tumor and B cells
hsa-miR-453046335654discovered in B
cells
hsa-miR-453146345655discovered in B
cells
hsa-miR-453246355656discovered in B
cells
hsa-miR-453346365657discovered in B
cells
hsa-miR-453446375658discovered in B
cells
hsa-miR-453546385659discovered in B
cells
hsa-miR-4536-3p46395660discovered in B
cells
hsa-miR-4536-5p46405661discovered in B
cells
hsa-miR-453746415662discovered in B
cells
hsa-miR-453846425663discovered in B
cells
hsa-miR-453946435664discovered in B
cells
hsa-miR-454046445665discovered in B
cells
hsa-miR-454-3p46455666embryoid body
cells, central
nevous system,
monocytes
hsa-miR-454-5p46465667embryoid body
cells, central
nevous system,
monocytes
hsa-miR-455-3p46475668basal cell
carcinoma, other
cancers
hsa-miR-455-5p46485669basal cell
carcinoma, other
cancers
hsa-miR-4632-3p46495670discovred in breast
tumor
hsa-miR-4632-5p46505671discovered in breast
tumor
hsa-miR-4633-3p46515672discovered in breast
tumor
hsa-miR-4633-5p46525673discovered in breast
tumor
hsa-miR-463446535674discovered in breast
tumor
hsa-miR-463546545675discovered in breast
tumor
hsa-miR-463646555676discovered in breast
tumor
hsa-miR-463746565677discovered in breast
tumor and
lymphoblastic
leukaemia
hsa-miR-4638-3p46575678discovered in breast
tumor
hsa-miR-4638-5p46585679discovered in breast
tumor
hsa-miR-4639-3p46595680discovered in breast
tumor
hsa-miR-4639-5p46605681discovered in breast
tumor
hsa-miR-4640-3p46615682discovered in breast
tumor
hsa-miR-4640-5p46625683discovered in breast
tumor
hsa-miR-464146635684discovered in breast
tumor
hsa-miR-464246645685discovered in breast
tumor
hsa-miR-464346655686discovered in breast
tumor
hsa-miR-464446665687discovered in breast
tumor
hsa-miR-4645-3p46675688discovered in breast
tumor
hsa-miR-4645-5p46685689discovered in breast
tumor
hsa-miR-4646-3p46695690discovered in breast
tumor
hsa-miR-4646-5p46705691discovered in breast
tumor
hsa-miR-464746715692discovered in breast
tumor
hsa-miR-464846725693discovered in breast
tumor
hsa-miR-4649-3p46735694discovered in breast
tumor
hsa-miR-4649-5p46745695discovered in breast
tumor
hsa-miR-4650-3p46755696discovered in breast
tumor
hsa-miR-4650-5p46765697discovered in breast
tumor
hsa-miR-465146775698discovered in breast
tumor
hsa-miR-4652-3p46785699discovered in breast
tumor
hsa-miR-4652-5p46795700discovered in breast
tumor
hsa-miR-4653-3p46805701discovered in breast
tumor
hsa-miR-4653-5p46815702discovered in breast
tumor
hsa-miR-465446825703discovered in breast
tumor
hsa-miR-4655-3p46835704discovered in breast
tumor
hsa-miR-4655-5p46845705discovered in breast
tumor
hsa-miR-465646855706discovered in breast
tumor
hsa-miR-465746865707discovered in breast
tumor
hsa-miR-465846875708discovered in breast
tumor
hsa-miR-4659a-3p46885709discovered in breast
tumor
hsa-miR-4659a-5p46895710discovered in breast
tumor
hsa-miR-4659b-3p46905711discovered in breast
tumor
hsa-miR-4659b-5p46915712discovered in breast
tumor
hsa-miR-46646925713
hsa-miR-466046935714discovered in breast
tumor
hsa-miR-4661-3p46945715discovered in breast
tumor
hsa-miR-4661-5p46955716discovered in breast
tumor
hsa-miR-4662a-3p46965717discovered in breast
tumor, psoriasis
hsa-miR-4662a-5p46975718discovered in breast
tumor, psoriasis
hsa-miR-4662b46985719discovered in breast
tumor
hsa-miR-466346995720discovered in breast
tumor
hsa-miR-4664-3p47005721discovered in breast
tumor
hsa-miR-4664-5p47015722discovered in breast
tumor
hsa-miR-4665-3p47025723discovered in breast
tumor
hsa-miR-4665-5p47035724discovered in breast
tumor
hsa-miR-4666a-3p47045725discovered in breast
tumor
hsa-miR-4666a-5p47055726discovered in breast
tumor
hsa-miR-4666b47065727
hsa-miR-4667-3p47075728discovered in breast
tumor
hsa-miR-4667-5p47085729discovered in breast
tumor
hsa-miR-4668-3p47095730discovered in breast
tumor
hsa-miR-4668-5p47105731discovered in breast
tumor
hsa-miR-466947115732discovered in breast
tumor
hsa-miR-4670-3p47125733discovered in breast
tumor
hsa-miR-4670-5p47135734discovered in breast
tumor
hsa-miR-4671-3p47145735discovered in breast
tumor
hsa-miR-4671-5p47155736discovered in breast
tumor
hsa-miR-467247165737discovered in breast
tumor
hsa-miR-467347175738discovered in breast
tumor
hsa-miR-467447185739discovered in breast
tumor
hsa-miR-467547195740discovered in breast
tumor
hsa-miR-4676-3p47205741discovered in breast
tumor
hsa-miR-4676-5p47215742discovered in breast
tumor
hsa-miR-4677-3p47225743discovered in breast
tumor, psoriasis
hsa-miR-4677-5p47235744discovered in breast
tumor, psoriasis
hsa-miR-467847245745discovered in breast
tumor
hsa-miR-467947255746discovered in breast
tumor
hsa-miR-4680-3p47265747discovered in breast
tumor
hsa-miR-4680-5p47275748discovered in breast
tumor
hsa-miR-468147285749discovered in breast
tumor
hsa-miR-468247295750discovered in breast
tumor
hsa-miR-468347305751discovered in breast
tumor
hsa-miR-4684-3p47315752discovered in breast
tumor
hsa-miR-4684-5p47325753discovered in breast
tumor
hsa-miR-4685-3p47335754discovered in breast
tumor
hsa-miR-4685-5p47345755discovered in breast
tumor
hsa-miR-468647355756discovered in breast
tumor
hsa-miR-4687-3p47365757discovered in breast
tumor
hsa-miR-4687-5p47375758discovered in breast
tumor
hsa-miR-468847385759discovered in breast
tumor
hsa-miR-468947395760discovered in breast
tumor
hsa-miR-4690-3p47405761discovered in breast
tumor
hsa-miR-4690-5p47415762discovered in breast
tumor
hsa-miR-4691-3p47425763discovered in breast
tumor
hsa-miR-4691-5p47435764discovered in breast
tumor
hsa-miR-469247445765discovered in breast
tumor
hsa-miR-4693-3p47455766discovered in breast
tumor
hsa-miR-4693-5p47465767discovered in breast
tumor
hsa-miR-4694-3p47475768discovered in breast
tumor
hsa-miR-4694-5p47485769discovered in breast
tumor
hsa-miR-4695-3p47495770discovered in breast
tumor
hsa-miR-4695-5p47505771discovered in breast
tumor
hsa-miR-469647515772discovered in breast
tumor
hsa-miR-4697-3p47525773discovered in breast
tumor
hsa-miR-4697-5p47535774discovered in breast
tumor
hsa-miR-469847545775discovered in breast
tumor
hsa-miR-4699-3p47555776discovered in breast
tumor
hsa-miR-4699-5p47565777discovered in breast
tumor
hsa-miR-4700-3p47575778discovered in breast
tumor
hsa-miR-4700-5p47585779discovered in breast
tumor
hsa-miR-4701-3p47595780discovered in breast
tumor
hsa-miR-4701-5p47605781discovered in breast
tumor
hsa-miR-4703-3p47615782discovered in breast
tumor
hsa-miR-4703-5p47625783discovered in breast
tumor
hsa-miR-4704-3p47635784discovered in breast
tumor
hsa-miR-4704-5p47645785discovered in breast
tumor
hsa-miR-470547655786discovered in breast
tumor
hsa-miR-470647665787discovered in breast
tumor
hsa-miR-4707-3p47675788discovered in breast
tumor
hsa-miR-4707-5p47685789discovered in breast
tumor
hsa-miR-4708-3p47695790discovered in breast
tumor
hsa-miR-4708-5p47705791discovered in breast
tumor
hsa-miR-4709-3p47715792discovered in breast
tumor
hsa-miR-4709-5p47725793discovered in breast
tumor
hsa-miR-471047735794discovered in breast
tumor
hsa-miR-4711-3p47745795discovered in breast
tumor
hsa-miR-4711-5p47755796discovered in breast
tumor
hsa-miR-4712-3p47765797discovered in breast
tumor
hsa-miR-4712-5p47775798discovered in breast
tumor
hsa-miR-4713-3p47785799discovered in breast
tumor
hsa-miR-4713-5p47795800discovered in breast
tumor
hsa-miR-4714-3p47805801discovered in breast
tumor
hsa-miR-4714-5p47815802discovered in breast
tumor
hsa-miR-4715-3p47825803discovered in breast
tumor
hsa-miR-4715-5p47835804discovered in breast
tumor
hsa-miR-4716-3p47845805discovered in breast
tumor
hsa-miR-4716-5p47855806discovered in breast
tumor
hsa-miR-4717-3p47865807discovered in breast
tumor
hsa-miR-4717-5p47875808discovered in breast
tumor
hsa-miR-471847885809discovered in breast
tumor
hsa-miR-471947895810discovered in breast
tumor
hsa-miR-4720-3p47905811discovered in breast
tumor
hsa-miR-4720-5p47915812discovered in breast
tumor
hsa-miR-472147925813discovered in breast
tumor
hsa-miR-4722-3p47935814discovered in breast
tumor
hsa-miR-4722-5p47945815discovered in breast
tumor
hsa-miR-4723-3p47955816discovered in breast
tumor
hsa-miR-4723-5p47965817discovered in breast
tumor
hsa-miR-4724-3p47975818discovered in breast
tumor
hsa-miR-4724-5p47985819discovered in breast
tumor
hsa-miR-4725-3p47995820discovered in breast
tumor
hsa-miR-4725-5p48005821discovered in breast
tumor
hsa-miR-4726-3p48015822discovered in breast
tumor
hsa-miR-4726-5p48025823discovered in breast
tumor
hsa-miR-4727-3p48035824discovered in breast
tumor
hsa-miR-4727-5p48045825discovered in breast
tumor
hsa-miR-4728-3p48055826discovered in breast
tumor
hsa-miR-4728-5p48065827discovered in breast
tumor
hsa-miR-472948075828discovered in breast
tumor
hsa-miR-473048085829discovered in breast
tumor
hsa-miR-4731-3p48095830discovered in breast
tumor
hsa-miR-4731-5p48105831discovered in breast
tumor
hsa-miR-4732-3p48115832discovered in breast
tumor
hsa-miR-4732-5p48125833discovered in breast
tumor
hsa-miR-4733-3p48135834discovered in breast
tumor
hsa-miR-4733-5p48145835discovered in breast
tumor
hsa-miR-473448155836discovered in breast
tumor
hsa-miR-4735-3p48165837discovered in breast
tumor
hsa-miR-4735-5p48175838discovered in breast
tumor
hsa-miR-473648185839discovered in breast
tumor
hsa-miR-473748195840discovered in breast
tumor
hsa-miR-4738-3p48205841discovered in breast
tumor
hsa-miR-4738-5p48215842discovered in breast
tumor
hsa-miR-473948225843discovered in breast
tumor
hsa-miR-4740-3p48235844discovered in breast
tumor
hsa-miR-4740-5p48245845discovered in breast
tumor
hsa-miR-474148255846discovered in breast
tumor, psoriasis
hsa-miR-4742-3p48265847discovered in breast
tumor, psoriasis
hsa-miR-4742-5p48275848discovered in breast
tumor
hsa-miR-4743-3p48285849discovered in breast
tumor
hsa-miR-4743-5p48295850discovered in breast
tumor
hsa-miR-474448305851discovered in breast
tumor
hsa-miR-4745-3p48315852discovered in breast
tumor
hsa-miR-4745-5p48325853discovered in breast
tumor
hsa-miR-4746-3p48335854discovered in breast
tumor
hsa-miR-4746-5p48345855discovered in breast
tumor
hsa-miR-4747-3p48355856discovered in breast
tumor
hsa-miR-4747-5p48365857discovered in breast
tumor
hsa-miR-474848375858discovered in breast
tumor
hsa-miR-4749-3p48385859discovered in breast
tumor
hsa-miR-4749-5p48395860discovered in breast
tumor
hsa-miR-4750-3p48405861discovered in breast
tumor
hsa-miR-4750-5p48415862discovered in breast
tumor
hsa-miR-475148425863discovered in breast
tumor
hsa-miR-475248435864discovered in breast
tumor
hsa-miR-4753-3p48445865discovered in breast
tumor
hsa-miR-4753-5p48455866discovered in breast
tumor
hsa-miR-475448465867discovered in breast
tumor
hsa-miR-4755-3p48475868discovered in breast
tumor
hsa-miR-4755-5p48485869discovered in breast
tumor
hsa-miR-4756-3p48495870discovered in breast
tumor
hsa-miR-4756-5p48505871discovered in breast
tumor
hsa-miR-4757-3p48515872discovered in breast
tumor
hsa-miR-4757-5p48525873discovered in breast
tumor
hsa-miR-4758-3p48535874discovered in breast
tumor
hsa-miR-4758-5p48545875discovered in breast
tumor
hsa-miR-475948555876discovered in breast
tumor
hsa-miR-4760-3p48565877discovered in breast
tumor
hsa-miR-4760-5p48575878discovered in breast
tumor
hsa-miR-4761-3p48585879discovered in breast
tumor
hsa-miR-4761-5p48595880discovered in breast
tumor
hsa-miR-4762-3p48605881discovered in breast
tumor
hsa-miR-4762-5p48615882discovered in breast
tumor
hsa-miR-4763-3p48625883discovered in breast
tumor
hsa-miR-4763-5p48635884discovered in breast
tumor
hsa-miR-4764-3p48645885discovered in breast
tumor
hsa-miR-4764-5p48655886discovered in breast
tumor
hsa-miR-476548665887discovered in breast
tumor
hsa-miR-4766-3p48675888discovered in breast
tumor
hsa-miR-4766-5p48685889discovered in breast
tumor
hsa-miR-476748695890discovered in breast
tumor
hsa-miR-4768-3p48705891discovered in breast
tumor
hsa-miR-4768-5p48715892discovered in breast
tumor
hsa-miR-4769-3p48725893discovered in breast
tumor
hsa-miR-4769-5p48735894discovered in breast
tumor
hsa-miR-477048745895discovered in breast
tumor
hsa-miR-477148755896discovered in breast
tumor
hsa-miR-4772-3p48765897discovered in breastenergy
tumor, bloodmetabolism/
monoclear cellsobesity
hsa-miR-4772-5p48775898discovered in breastenergy
tumor, bloodmetabolism/
monoclear cellsobesity
hsa-miR-477348785899discovered in breast
tumor
hsa-miR-4774-3p48795900discovered in breast
tumor and
Lymphoblastic
leukemia
hsa-miR-4774-5p48805901discovered in breast
tumor and
Lymphoblastic
leukemia
hsa-miR-477548815902discovered in breast
tumor
hsa-miR-4776-3p48825903discovered in breast
tumor
hsa-miR-4776-5p48835904discovered in breast
tumor
hsa-miR-4777-3p48845905discovered in breast
tumor
hsa-miR-4777-5p48855906discovered in breast
tumor
hsa-miR-4778-3p48865907discovered in breast
tumor
hsa-miR-4778-5p48875908discovered in breast
tumor
hsa-miR-477948885909discovered in breast
tumor
hsa-miR-478048895910discovered in breast
tumor
hsa-miR-4781-3p48905911discovered in breast
tumor
hsa-miR-4781-5p48915912discovered in breast
tumor
hsa-miR-4782-3p48925913discovered in breast
tumor
hsa-miR-4782-5p48935914discovered in breast
tumor
hsa-miR-4783-3p48945915discovered in breast
tumor
hsa-miR-4783-5p48955916discovered in breast
tumor
hsa-miR-478448965917discovered in breast
tumor
hsa-miR-478548975918discovered in breast
tumor
hsa-miR-4786-3p48985919discovered in breast
tumor
hsa-miR-4786-5p48995920discovered in breast
tumor
hsa-miR-4787-3p49005921discovered in breast
tumor
hsa-miR-4787-5p49015922discovered in breast
tumor
hsa-miR-478849025923discovered in breast
tumor
hsa-miR-4789-3p49035924discovered in breast
tumor
hsa-miR-4789-5p49045925discovered in breast
tumor
hsa-miR-4790-3p49055926discovered in breast
tumor
hsa-miR-4790-5p49065927discovered in breast
tumor
hsa-miR-479149075928discovered in breast
tumor
hsa-miR-479249085929discovered in breast
tumor
hsa-miR-4793-3p49095930discovered in breast
tumor
hsa-miR-4793-5p49105931discovered in breast
tumor
hsa-miR-479449115932discovered in breast
tumor
hsa-miR-4795-3p49125933discovered in breast
tumor
hsa-miR-4795-5p49135934discovered in breast
tumor
hsa-miR-4796-3p49145935discovered in breast
tumor
hsa-miR-4796-5p49155936discovered in breast
tumor
hsa-miR-4797-3p49165937discovered in breast
tumor
hsa-miR-4797-5p49175938discovered in breast
tumor
hsa-miR-4798-3p49185939discovered in breast
tumor
hsa-miR-4798-5p49195940discovered in breast
tumor
hsa-miR-4799-3p49205941discovered in breast
tumor
hsa-miR-4799-5p49215942discovered in breast
tumor
hsa-miR-4800-3p49225943discovered in breast
tumor
hsa-miR-4800-5p49235944discovered in breast
tumor
hsa-miR-480149245945discovered in breast
tumor
hsa-miR-4802-3p49255946discovered in breast
tumor, psoriasis
hsa-miR-4802-5p49265947discovered in breast
tumor, psoriasis
hsa-miR-480349275948discovered in breast
tumor
hsa-miR-4804-3p49285949discovered in breast
tumor
hsa-miR-4804-5p49295950discovered in breast
tumor
hsa-miR-483-3p49305951aderonocorticaloncogenic
carcinoma,
rectal/pancreatic
cancer,
proliferation of
wounded
epithelial cells
hsa-miR-483-5p49315952cartilageaderonocorticalangiogenesis
(chondrocyte), fetalcarcinoma
brain
hsa-miR-48449325953mitochondrial
network
hsa-miR-485-3p49335954
hsa-miR-485-5p49345955ovarian epithelial
tumor
hsa-miR-486-3p49355956erythroid cellsvarious cancers
hsa-miR-486-5p49365957stem cells (adipose)various cancers
hsa-miR-487a49375958laryngeal
carcinoma
hsa-miR-487b49385959neuroblastoma, pulmonary
carcinogenesis
hsa-miR-488-3p49395960prostate cancer,
others
hsa-miR-488-5p49405961prostate cancer,
others
hsa-miR-48949415962mesenchymal stemosteogenesis
cells
hsa-miR-490-3p49425963neuroblastoma,
terine leiomyoma
(ULM)/muscle
hsa-miR-490-5p49435964neuroblastoma,
terine leiomyoma
(ULM)/muscle
hsa-miR-491-3p49445965various cancers,pro-apoptosis
brain disease
hsa-miR-491-5p49455966various cancers,pro-apoptosis
brain disease
hsa-miR-49249465967
hsa-miR-493-3p49475968myeloid cells,
pancreas (islet)
hsa-miR-493-5p49485969myeloid cells,
pancreas (islet)
hsa-miR-49449495970epithelial cellsvarious cancerscell cycle
hsa-miR-495-3p49505971plateletvarious cancers
(gastric, MLL
leukemia,
pancreatic etc)
and inflammation
hsa-miR-495-5p49515972plateletvarious cancers
(gastric, MLL
leukemia,
pancreatic etc)
and inflammation
hsa-miR-49649525973Blood
hsa-miR-497-3p49535974various cancerstumor
(breast,supressor/pro-
colorectal, etc)apoptosis
hsa-miR-497-5p49545975various cancerstumor
(breast,supressor/pro-
colorectal, etc)apoptosis
hsa-miR-49849555976autoimmuno (e.g.
rheumatoid
arthritis)
hsa-miR-4999-3p49565977
hsa-miR-4999-5p49575978
hsa-miR-499a-3p49585979heart, cardiac stemcardiovascularcardiomyocyte
cellsdiseasedifferentiation
hsa-miR-499a-5p49595980heart, cardiac stemcardiovascularcardiomyocyte
cellsdiseasedifferentiation
hsa-miR-499b-3p49605981heart, cardiac stemcardiovascularcardiomyocyte
cellsdiseasedifferentiation
hsa-miR-499b-5p49615982heart, cardiac stemcardiovascularcardiomyocyte
cellsdiseasedifferentiation
hsa-miR-5000-3p49625983discovered in
lymphoblastic
leukaemia
hsa-miR-5000-5p49635984discovered in
lymphoblastic
leukaemia
hsa-miR-5001-3p49645985
hsa-miR-5001-5p49655986
hsa-miR-5002-3p49665987
hsa-miR-5002-5p49675988
hsa-miR-5003-3p49685989
hsa-miR-5003-5p49695990
hsa-miR-5004-3p49705991
hsa-miR-5004-5p49715992
hsa-miR-5006-3p49725993discovered in
lymphoblastic
leukaemia
hsa-miR-5006-5p49735994discovered in
lymphoblastic
leukaemia
hsa-miR-5007-3p49745995
hsa-miR-5007-5p49755996
hsa-miR-5008-3p49765997
hsa-miR-5008-5p49775998
hsa-miR-5009-3p49785999
hsa-miR-5009-5p49796000
hsa-miR-500a-3p49806001
hsa-miR-500a-5p49816002
hsa-miR-500b49826003Blood (plasma)
hsa-miR-5010-3p49836004abnormal skin
(psoriasis)
hsa-miR-5010-5p49846005abnormal skin
(psoriasis)
hsa-miR-5011-3p49856006
hsa-miR-5011-5p49866007
hsa-miR-501-3p49876008
hsa-miR-501-5p49886009
hsa-miR-502-3p49896010various cancers
(hepatocellular,
ovarian, breast)
hsa-miR-502-5p49906011various cancers
(hepatocellular,
ovarian, breast)
hsa-miR-503-3p49916012ovary
hsa-miR-503-5p49926013ovary
hsa-miR-50449936014glioblastoma
hsa-miR-504749946015
hsa-miR-505-3p49956016breast cancer
hsa-miR-505-5p49966017breast cancer
hsa-miR-506-3p49976018various cancers
hsa-miR-506-5p49986019various cancers
hsa-miR-50749996020
hsa-miR-508-3p50006021renal cell
carcinoma
hsa-miR-508-5p50016022endothelial
progenitor cells
(EPCs)
hsa-miR-508750026023
hsa-miR-508850036024
hsa-miR-5089-3p50046025
hsa-miR-5089-5p50056026
hsa-miR-509050066027
hsa-miR-509150076028
hsa-miR-509250086029
hsa-miR-509350096030
hsa-miR-509-3-5p50106031testis
hsa-miR-509-3p50116032renal cell
carcinoma, brain
disease
hsa-miR-509450126033
hsa-miR-509550136034cervical cancer
hsa-miR-509-5p50146035metabolic
syndrome, brain
disease
hsa-miR-509650156036cervical cance
hsa-miR-51050166037brain
hsa-miR-510050176038discoverd in
Salivary gland
hsa-miR-51150186039dendritic cells and
macrophages
hsa-miR-512-3p50196040embryonic stem
cells, placenta
hsa-miR-512-5p50206041embryonic stem
cells, placenta,
hsa-miR-513a-3p50216042lung carcinoma
hsa-miR-513a-5p50226043endothelial cells
hsa-miR-513b50236044follicular
lymphoma
hsa-miR-513c-3p50246045
hsa-miR-513c-5p50256046
hsa-miR-514a-3p50266047
hsa-miR-514a-5p50276048
hsa-miR-514b-3p50286049various cancer
cells
hsa-miR-514b-5p50296050various cancer
cells
hsa-miR-515-3p50306051
hsa-miR-515-5p50316052placenta
hsa-miR-516a-3p50326053frontal cortex
hsa-miR-516a-5p50336054placenta
hsa-miR-516b-3p50346055
hsa-miR-516b-5p50356056
hsa-miR-517-5p50366057placenta
hsa-miR-517a-3p50376058placenta
hsa-miR-517b-3p50386059placenta
hsa-miR-517c-3p50396060placenta
hsa-miR-518650406061discovered in
lymphoblastic
leukaemia
hsa-miR-5187-3p50416062discovered in
lymphoblastic
leukaemia, skin
(psoriasis)
hsa-miR-5187-5p50426063discovered in
lymphoblastic
leukaemia, skin
(psoriasis)
hsa-miR-518850436064discovered in
lymphoblastic
leukaemia
hsa-miR-518950446065discovered in
lymphoblastic
leukaemia
hsa-miR-518a-3p50456066HCC
hsa-miR-518a-5p50466067various cancer
cells
hsa-miR-518b50476068placentaHCCcell cycle
progression
hsa-miR-518c-3p50486069placenta
hsa-miR-518c-5p50496070placenta
hsa-miR-518d-3p50506071
hsa-miR-518d-5p50516072
hsa-miR-518e-3p50526073HCCcell cycle
progression
hsa-miR-518e-5p50536074HCCcell cycle
progression
hsa-miR-518f-3p50546075placenta
hsa-miR-518f-5p50556076placenta
hsa-miR-519050566077discovered in
lymphoblastic
leukaemia
hsa-miR-519150576078discovered in
lymphoblastic
leukaemia
hsa-miR-519250586079discovered in
lymphoblastic
leukaemia
hsa-miR-519350596080discovered in
lymphoblastic
leukaemia
hsa-miR-519450606081discovered in
lymphoblastic
leukaemia
hsa-miR-5195-3p50616082discovered in
lymphoblastic
leukaemia
hsa-miR-5195-5p50626083discovered in
lymphoblastic
leukaemia
hsa-miR-5196-3p50636084discovered in
lymphoblastic
leukaemia
hsa-miR-5196-5p50646085discovered in
lymphoblastic
leukaemia
hsa-miR-5197-3p50656086discovered in
lymphoblastic
leukaemia
hsa-miR-5197-5p50666087discovered in
lymphoblastic
leukaemia
hsa-miR-519a-3p50676088placentaHCC
hsa-miR-519a-5p50686089placentaHCC
hsa-miR-519b-3p50696090breast cancer
hsa-miR-519b-5p50706091breast cancer
hsa-miR-519c-3p50716092
hsa-miR-519c-5p50726093
hsa-miR-519d50736094placenta
hsa-miR-519e-3p50746095placenta
hsa-miR-519e-5p50756096placenta
hsa-miR-520a-3p50766097placenta
hsa-miR-520a-5p50776098placenta
hsa-miR-520b50786099breast cancer
hsa-miR-520c-3p50796100gastric cancer,
breast tumor
hsa-miR-520c-5p50806101breast tumor
hsa-miR-520d-3p50816102various cancer
cells
hsa-miR-520d-5p50826103various cancer
cells
hsa-miR-520e50836104hepatomatomor
suppressor
hsa-miR-520f50846105breast cancer
hsa-miR-520g50856106HCC, bladder
cancer, breast
cancer
hsa-miR-520h50866107placental specific
hsa-miR-52150876108prostate cancer
hsa-miR-522-3p50886109HCC
hsa-miR-522-5p50896110HCC
hsa-miR-523-3p50906111
hsa-miR-523-5p50916112
hsa-miR-524-3p50926113colon cancer stem
cells
hsa-miR-524-5p50936114placental specificgliomas
hsa-miR-525-3p50946115placental specificHCC
hsa-miR-525-5p50956116placental specific
hsa-miR-526a50966117placental specific
hsa-miR-526b-3p50976118placental specific
hsa-miR-526b-5p50986119placental specific
hsa-miR-52750996120
hsa-miR-532-3p51006121ALL
hsa-miR-532-5p51016122ALL
hsa-miR-539-3p51026123
hsa-miR-539-5p51036124
hsa-miR-541-3p51046125
hsa-miR-541-5p51056126
hsa-miR-542-3p51066127monocytes
hsa-miR-542-5p51076128basal cell
carcinoma,
neuroblastoma
hsa-miR-54351086129
hsa-miR-544a51096130osteocarcoma
hsa-miR-544b51106131osteocarcoma
hsa-miR-545-3p51116132
hsa-miR-545-5p51126133rectal cancer
hsa-miR-54851136134
hsa-miR-548-3p51146135
hsa-miR-548-5p51156136
hsa-miR-548a51166137identified in
colorectal
microRNAome
hsa-miR-548a-3p51176138identified in
colorectal
microRNAome
hsa-miR-548a-5p51186139identified in
colorectal
microRNAome
hsa-miR-548aa51196140identified in
cervical tumor
hsa-miR-548ab51206141discovered in B-
cells
hsa-miR-548ac51216142discovered in B-
cells
hsa-miR-548ad51226143discovered in B-
cells
hsa-miR-548ae51236144discovered in B-
cells
hsa-miR-548ag51246145discovered in B-
cells
hsa-miR-548ah-3p51256146discovered in B-
cells
hsa-miR-548ah-5p51266147discovered in B-
cells
hsa-miR-548ai51276148discovered in B-
cells
hsa-miR-548aj-3p51286149discovered in B-
cells
hsa-miR-548aj-5p51296150discovered in B-
cells
hsa-miR-548ak51306151discovered in B-
cells
hsa-miR-548al51316152discovered in B-
cells
hsa-miR-548am-3p51326153discovered in B-
cells
hsa-miR-548am-5p51336154discovered in B-
cells
hsa-miR-548an51346155discovered in B-
cells
hsa-miR-548ao-3p51356156
hsa-miR-548ao-5p51366157
hsa-miR-548ap-3p51376158
hsa-miR-548ap-5p51386159
hsa-miR-548aq-3p51396160
hsa-miR-548aq-5p51406161
hsa-miR-548ar-3p51416162
hsa-miR-548ar-5p51426163
hsa-miR-548as-3p51436164
hsa-miR-548as-5p51446165
hsa-miR-548at-3p51456166prostate cancer
hsa-miR-548at-5p51466167prostate cancer
hsa-miR-548au-3p51476168
hsa-miR-548au-5p51486169
hsa-miR-548av-3p51496170
hsa-miR-548av-5p51506171
hsa-miR-548aw51516172prostate cancer
hsa-miR-548ay-3p51526173discovered in
abnormal skin
(psoriasis)
hsa-miR-548ay-5p51536174discovered in
abnormal skin
(psoriasis)
hsa-miR-548az-3p51546175discovered in
abnormal skin
(psoriasis)
hsa-miR-548az-5p51556176discovered in
abnormal skin
(psoriasis)
hsa-miR-548b-3p51566177identified in
colorectal
microRNAome
hsa-miR-548b-5p51576178immune cells,
frontal cortex
hsa-miR-548c-3p51586179identified in
colorectal
microRNAome
hsa-miR-548c-5p51596180immune cells,
frontal cortex
hsa-miR-548d-3p51606181identified in
colorectal
microRNAome
hsa-miR-548d-5p51616182identified in
colorectal
microRNAome
hsa-miR-548e51626183embryonic stem
cells
hsa-miR-548f51636184embryonic stem
cells
hsa-miR-548g-3p51646185embryonic stem
cells
hsa-miR-548g-5p51656186embryonic stem
cells
hsa-miR-548h-3p51666187embryonic stem
cells
hsa-miR-548h-5p51676188embryonic stem
cells
hsa-miR-548i51686189embryonic stem
cells, immune cells
hsa-miR-548j51696190immune cells
hsa-miR-548k51706191embryonic stem
cells
hsa-miR-548151716192embryonic stem
cells
hsa-miR-548m51726193embryonic stem
cells
hsa-miR-548n51736194embryonic stem
cells, immune cells
hsa-miR-548o-3p51746195embryonic stem
cells
hsa-miR-548o-5p51756196embryonic stem
cells
hsa-miR-548p51766197embryonic stem
cells
hsa-miR-548q51776198ovarian cancer
cells
hsa-miR-548s51786199discovered in the
melanoma
MicroRNAome
hsa-miR-548t-3p51796200discovered in the
melanoma
MicroRNAome
hsa-miR-548t-5p51806201discovered in the
melanoma
MicroRNAome
hsa-miR-548u51816202discovered in the
melanoma
MicroRNAome
hsa-miR-548w51826203discovered in the
melanoma
MicroRNAome
hsa-miR-548y51836204
hsa-miR-548z51846205discovered in
cervical tumor
hsa-miR-549a51856206discovered in a
colorectal
MicroRNAome
hsa-miR-550a-3-5p51866207Hepatocellular
Carcinoma
hsa-miR-550a-3p51876208Hepatocellular
Carcinoma
hsa-miR-550a-5p51886209Hepatocellular
Carcinoma
hsa-miR-550b-2-5p51896210discovered in
cervical tumor
hsa-miR-550b-3p51906211discovered in
cervical tumor
hsa-miR-551a51916212gastric cancer
hsa-miR-551b-3p51926213hepatocytes
hsa-miR-551b-5p51936214hepatocytes
hsa-miR-55251946215discovered in a
colorectal
MicroRNAome
hsa-miR-55351956216discovered in a
colorectal
MicroRNAome
hsa-miR-55451966217discovered in a
colorectal
MicroRNAome
hsa-miR-55551976218discovered in a
colorectal
MicroRNAome
hsa-miR-556-3p51986219discovered in a
colorectal
MicroRNAome
hsa-miR-556-5p51996220discovered in a
colorectal
MicroRNAome
hsa-miR-55752006221liver (hepatocytes)
hsa-miR-5571-3p52016222discoveredd in
Salivary gland
hsa-miR-5571-5p52026223discoveredd in
Salivary gland
hsa-miR-557252036224discoveredd in
Salivary gland
hsa-miR-5579-3p52046225
hsa-miR-5579-5p52056226
hsa-miR-55852066227neuroblastoma
hsa-miR-5580-3p52076228
hsa-miR-5580-5p52086229
hsa-miR-5581-3p52096230
hsa-miR-5581-5p52106231
hsa-miR-5582-3p52116232
hsa-miR-5582-5p52126233
hsa-miR-5583-3p52136234
hsa-miR-5583-5p52146235
hsa-miR-5584-3p52156236
hsa-miR-5584-5p52166237
hsa-miR-5585-3p52176238
hsa-miR-5585-5p52186239
hsa-miR-5586-3p52196240
hsa-miR-5586-5p52206241
hsa-miR-5587-3p52216242
hsa-miR-5587-5p52226243
hsa-miR-5588-3p52236244
hsa-miR-5588-5p52246245
hsa-miR-5589-3p52256246
hsa-miR-5589-5p52266247
hsa-miR-55952276248
hsa-miR-5590-3p52286249
hsa-miR-5590-5p52296250
hsa-miR-5591-3p52306251
hsa-miR-5591-5p52316252
hsa-miR-561-3p52326253multiple myeloma
hsa-miR-561-5p52336254multiple myeloma
hsa-miR-56252346255
hsa-miR-56352356256discovered in a
colorectal
MicroRNAome
hsa-miR-56452366257Chronic myeloid
leukemia
hsa-miR-56652376258MALT
lymphoma/lymphocyte
hsa-miR-56752386259colorectal cancer
hsa-miR-56852396260discovered in a
colorectal
MicroRNAome
hsa-miR-568052406261Associated with
metastatic
prostate cancer
hsa-miR-5681a52416262Associated with
metastatic
prostate cancer
hsa-miR-5681b52426263Associated with
metastatic
prostate cancer
hsa-miR-568252436264Associated with
metastatic
prostate cancer
hsa-miR-568352446265Associated with
metastatic
prostate cancer
hsa-miR-568452456266Associated with
metastatic
prostate cancer
hsa-miR-568552466267Associated with
metastatic
prostate cancer
hsa-miR-568652476268Associated with
metastatic
prostate cancer
hsa-miR-568752486269Associated with
metastatic
prostate cancer
hsa-miR-568852496270Associated with
metastatic
prostate cancer
hsa-miR-568952506271Associated with
metastatic
prostate cancer
hsa-miR-56952516272
hsa-miR-569052526273Associated with
metastatic
prostate cancer
hsa-miR-569152536274Associated with
metastatic
prostate cancer
hsa-miR-5692a52546275Associated with
metastatic
prostate cancer
hsa-miR-5692b52556276Associated with
metastatic
prostate cancer
hsa-miR-5692c52566277Associated with
metastatic
prostate cancer
hsa-miR-569352576278Associated with
metastatic
prostate cancer
hsa-miR-569452586279Associated with
metastatic
prostate cancer
hsa-miR-569552596280Associated with
metastatic
prostate cancer
hsa-miR-569652606281Associated with
metastatic
prostate cancer
hsa-miR-569752616282Associated with
metastatic
prostate cancer
hsa-miR-569852626283Associated with
metastatic
prostate cancer
hsa-miR-569952636284Associated with
metastatic
prostate cancer
hsa-miR-570052646285Associated with
metastatic
prostate cancer
hsa-miR-570152656286Associated with
metastatic
prostate cancer
hsa-miR-570252666287Associated with
metastatic
prostate cancer
hsa-miR-570352676288Associated with
metastatic
prostate cancer
hsa-miR-570-3p52686289follicular
lymphoma
hsa-miR-570452696290Associated with
metastatic
prostate cancer
hsa-miR-570552706291Associated with
metastatic
prostate cancer
hsa-miR-570-5p52716292follicular
lymphoma
hsa-miR-570652726293Associated with
metastatic
prostate cancer
hsa-miR-570752736294Associated with
metastatic
prostate cancer
hsa-miR-570852746295Associated with
metastatic
prostate cancer
hsa-miR-57152756296frontal cortex
hsa-miR-57252766297circulatingbasal cell
microRNA (incarcinoma
plasma)
hsa-miR-57352776298discovered in the
colorectal
MicroRNAome
hsa-miR-573952786299endothelial cells
hsa-miR-574-3p52796300blood (myeloidfollicular
cells)lymphoma
hsa-miR-574-5p52806301sem*n
hsa-miR-57552816302gastric cancer
hsa-miR-576-3p52826303discovered in a
colorectal
MicroRNAome
hsa-miR-576-5p52836304cartilage/
chondrocyte
hsa-miR-57752846305discovered in a
colorectal
MicroRNAome
hsa-miR-57852856306discovered in a
colorectal
MicroRNAome
hsa-miR-578752866307fibroblast
hsa-miR-57952876308
hsa-miR-58052886309breast cancer
hsa-miR-58152896310liver (hepatocytes)
hsa-miR-582-3p52906311cartilage/chondrocytebladder cancer
hsa-miR-582-5p52916312bladder cancer
hsa-miR-58352926313rectal cancer cells
hsa-miR-584-3p52936314tumor cells
(follicular
lymphoma, rectal
cancer cells)
hsa-miR-584-5p52946315tumor cells
(follicular
lymphoma, rectal
cancer cells)
hsa-miR-58552956316oral squamous
cell carcinoma
hsa-miR-58652966317discovered in a
colorectal
MicroRNAome
hsa-miR-58752976318discovered in a
colorectal
MicroRNAome
hsa-miR-58852986319discovered in a
colorectal
MicroRNAome
hsa-miR-589-3p52996320mesothelial cells
hsa-miR-589-5p53006321mesothelial cells
hsa-miR-590-3p53016322cardiomyocytesCell cycle
progression
hsa-miR-590-5p53026323cardiomyocytesCell cycle
progression
hsa-miR-59153036324neuroblastoma
hsa-miR-59253046325hepatocellular
carcinoma
hsa-miR-593-3p53056326esophageal cancer
hsa-miR-593-5p53066327esophageal cancer
hsa-miR-59553076328heart failure
hsa-miR-59653086329ependymoma,
cancers
hsa-miR-59753096330discovered in a
colorectal
MicroRNAome
hsa-miR-59853106331Blood
(lymphocytes)
hsa-miR-59953116332Multiple sclerosis
hsa-miR-60053126333discovered in a
colorectal
MicroRNAome
hsa-miR-60153136334various cancers
(colonrectal,
gastric)
hsa-miR-60253146335oocyte
hsa-miR-60353156336
hsa-miR-60453166337discovered in a
colorectal
MicroRNAome
hsa-miR-60553176338discovered in a
colorectal
MicroRNAome
hsa-miR-60653186339discovered in a
colorectal
MicroRNAome
hsa-miR-606853196340discovered in
endothelial cells
hsa-miR-606953206341discovered in
endothelial cells
hsa-miR-60753216342discovered in a
colorectal
MicroRNAome
hsa-miR-607053226343discovered in a
colorectal
MicroRNAome
hsa-miR-607153236344discovered in
endothelial cells
hsa-miR-607253246345discovered in
endothelial cells
hsa-miR-607353256346discovered in
endothelial cells
hsa-miR-607453266347discovered in
endothelial cells
hsa-miR-607553276348discovered in
endothelial cells
hsa-miR-607653286349discovered in
endothelial cells
hsa-miR-607753296350discovered in
endothelial cells
hsa-miR-607853306351discovered in
endothelial cells
hsa-miR-607953316352discovered in
endothelial cells
hsa-miR-60853326353various cancers
hsa-miR-608053336354discovered in
endothelial cells
hsa-miR-608153346355discovered in
endothelial cells
hsa-miR-608253356356discovered in
endothelial cells
hsa-miR-608353366357discovered in
endothelial cells
hsa-miR-608453376358discovered in
endothelial cells
hsa-miR-608553386359discovered in
endothelial cells
hsa-miR-608653396360embryonic stem
cells
hsa-miR-608753406361embryonic stem
cells
hsa-miR-608853416362embryonic stem
cells
hsa-miR-608953426363embryonic stem
cells
hsa-miR-60953436364discovered in a
colorectal
MicroRNAome
hsa-miR-609053446365embryonic stem
cells
hsa-miR-61053456366gastric cancer
hsa-miR-61153466367Renal cell
carcinoma
hsa-miR-61253476368AM leukemia
hsa-miR-612453486369
hsa-miR-612553496370
hsa-miR-612653506371
hsa-miR-612753516372
hsa-miR-612853526373
hsa-miR-612953536374
hsa-miR-61353546375lipid metabollism
hsa-miR-613053556376
hsa-miR-613153566377
hsa-miR-613253576378
hsa-miR-613353586379
hsa-miR-613453596380
hsa-miR-61453606381circulating
micrRNAs (in
Plasma)
hsa-miR-615-3p53616382
hsa-miR-615-5p53626383
hsa-miR-616-3p53636384prostate cancer
hsa-miR-616553646385Pro-apoptotic
factor
hsa-miR-616-5p53656386prostate cancer
hsa-miR-61753666387
hsa-miR-61853676388
hsa-miR-61953686389discovered in a
colorectal
MicroRNAome
hsa-miR-62053696390discovered in a
colorectal
MicroRNAome
hsa-miR-62153706391
hsa-miR-62253716392
hsa-miR-62353726393
hsa-miR-624-3p53736394chondrocyte
hsa-miR-624-5p53746395chondrocyte
hsa-miR-625-3p53756396liver (hepatocytes), circulatingvarious cancers
(blood)
hsa-miR-625-5p53766397liver (hepatocytes), circulatingvarious cancers
(blood)
hsa-miR-62653776398discovered in the
colorectal
MicroRNAome
hsa-miR-62753786399colorectal cancer
hsa-miR-628-3p53796400neuroblastoma
hsa-miR-628-5p53806401neuroblastoma
hsa-miR-629-3p53816402B-lineage ALL, T
cell lupus,
RCC/kidney
hsa-miR-629-5p53826403B-lineage ALL, T
cell lupus,
RCC/kidney
hsa-miR-63053836404chondrocytesrectal cancer
hsa-miR-63153846405discovered in the
colorectal
MicroRNAom
hsa-miR-63253856406myelodysplastic
syndromes
hsa-miR-63353866407multiple sclerosis
hsa-miR-63453876408cartilage/
chondrocyte
hsa-miR-63553886409discovered in the
colorectal
MicroRNAome
hsa-miR-63653896410myelodysplastic
syndromes
hsa-miR-63753906411discovered in the
colorectal
MicroRNAome
hsa-miR-63853916412Lupus nephritis,
basal cell
carcinoma
hsa-miR-63953926413discovered in the
colorectal
MicroRNAome
hsa-miR-64053936414Chronic
lymphocytic
leukemia
hsa-miR-64153946415cartilage/
chondrocyte
hsa-miR-642a-3p53956416adipocyte
hsa-miR-642a-5p53966417discovered in the
colorectal
MicroRNAome
hsa-miR-642b-3p53976418discovered in a
cervial tumo
hsa-miR-642b-5p53986419discovered in a
cervial tumo
hsa-miR-64353996420discovered in the
colorectal
MicroRNAome
hsa-miR-644a54006421
hsa-miR-64554016422ovarian cancer
hsa-miR-64654026423
hsa-miR-64754036424prostate and lung
cancer
hsa-miR-64854046425circulating
micrRNAs (in
Plasma)
hsa-miR-64954056426Serum
hsa-miR-6499-3p54066427discovered in
abnormal skin
(psoriasis)
hsa-miR-6499-5p54076428discovered in
abnormal skin
(psoriasis)
hsa-miR-65054086429melanoma
hsa-miR-6500-3p54096430discovered in
abnormal skin
(psoriasis)
hsa-miR-6500-5p54106431discovered in
abnormal skin
(psoriasis)
hsa-miR-6501-3p54116432discovered in
abnormal skin
(psoriasis)
hsa-miR-6501-5p54126433discovered in
abnormal skin
(psoriasis)
hsa-miR-6502-3p54136434discovered in
abnormal skin
(psoriasis)
hsa-miR-6502-5p54146435discovered in
abnormal skin
(psoriasis)
hsa-miR-6503-3p54156436discovered in
abnormal skin
(psoriasis)
hsa-miR-6503-5p54166437discovered in
abnormal skin
(psoriasis)
hsa-miR-6504-3p54176438discovered in
abnormal skin
(psoriasis)
hsa-miR-6504-5p54186439discovered in
abnormal skin
(psoriasis)
hsa-miR-6505-3p54196440discovered in
abnormal skin
(psoriasis)
hsa-miR-6505-5p54206441discovered in
abnormal skin
(psoriasis)
hsa-miR-6506-3p54216442discovered in
abnormal skin
(psoriasis)
hsa-miR-6506-5p54226443discovered in
abnormal skin
(psoriasis)
hsa-miR-6507-3p54236444discovered in
abnormal skin
(psoriasis)
hsa-miR-6507-5p54246445discovered in
abnormal skin
(psoriasis)
hsa-miR-6508-3p54256446discovered in
abnormal skin
(psoriasis)
hsa-miR-6508-5p54266447discovered in
abnormal skin
(psoriasis)
hsa-miR-6509-3p54276448discovered in
abnormal skin
(psoriasis)
hsa-miR-6509-5p54286449discovered in
abnormal skin
(psoriasis)
hsa-miR-65154296450discovered in thelung cancer
colorectal
MicroRNAome
hsa-miR-6510-3p54306451discovered in
abnormal skin
(psoriasis)
hsa-miR-6510-5p54316452discovered in
abnormal skin
(psoriasis)
hsa-miR-6511a-3p54326453discovered in
abnormal skin
(psoriasis) and
epididymis
hsa-miR-6511a-5p54336454discovered in
abnormal skin
(psoriasis) and
epididymis
hsa-miR-6511b-3p54346455discovered in
epididymis
hsa-miR-6511b-5p54356456discovered in
epididymis
hsa-miR-6512-3p54366457discovered in
abnormal skin
(psoriasis)
hsa-miR-6512-5p54376458discovered in
abnormal skin
(psoriasis)
hsa-miR-6513-3p54386459discovered in
abnormal skin
(psoriasis)
hsa-miR-6513-5p54396460discovered in
abnormal skin
(psoriasis)
hsa-miR-6514-3p54406461discovered in
abnormal skin
(psoriasis)
hsa-miR-6514-5p54416462discovered in
abnormal skin
(psoriasis)
hsa-miR-6515-3p54426463discovered in
abnormal skin
(psoriasis) and
epididymis
hsa-miR-6515-5p54436464discovered in
abnormal skin
(psoriasis) and
epididymis
hsa-miR-652-3p54446465rectal cancer cells
hsa-miR-652-5p54456466rectal cancer cells
hsa-miR-65354466467Discovered in the
colorectal
MicroRNAome
hsa-miR-654-3p54476468Discovered in the
colorectal
MicroRNAome
hsa-miR-654-5p54486469bone marrowprostate cancer
hsa-miR-65554496470
hsa-miR-65654506471various cancers
hsa-miR-65754516472oligodendrocytesdiabetes
hsa-miR-65854526473gastric cancer
hsa-miR-659-3p54536474myoblast
hsa-miR-659-5p54546475myoblast
hsa-miR-660-3p54556476myoblast
hsa-miR-660-5p54566477myoblast
hsa-miR-66154576478breast cancer
hsa-miR-66254586479endothelial
progenitor cells,
oocytes
hsa-miR-663a54596480follicular
lymphoma, Lupus
nephritis
hsa-miR-663b54606481follicular
lymphoma, Lupus
nephritis
hsa-miR-664a-3p54616482embryonic stemcomponent of
cellsSnoRNAs
hsa-miR-664a-5p54626483embryonic stemcomponent of
cellsSnoRNAs
hsa-miR-664b-3p54636484embryonic stemcomponent of
cellsSnoRNAs
hsa-miR-664b-5p54646485embryonic stemcomponent of
cellsSnoRNAs
hsa-miR-66554656486breast cancer
hsa-miR-66854666487keratinocytessenescence
hsa-miR-67054676488
hsa-miR-671-3p54686489
hsa-miR-6715a-3p54696490discovered in
epididymis
hsa-miR-6715b-3p54706491discovered in
epididymis
hsa-miR-6715b-5p54716492discovered in
epididymis
hsa-miR-671-5p54726493rectal cancer,
prolactinomas
hsa-miR-6716-3p54736494discovered in
epididymis
hsa-miR-6716-5p54746495discovered in
epididymis
hsa-miR-6717-5p54756496discovered in
epididymis
hsa-miR-6718-5p54766497discovered in
epididymis
hsa-miR-6719-3p54776498discovered in
epididymis
hsa-miR-6720-3p54786499discovered in
epididymis
hsa-miR-6721-5p54796500discovered in
epididymis
hsa-miR-6722-3p54806501discovered in
epididymis
hsa-miR-6722-5p54816502discovered in
epididymis
hsa-miR-6723-5p54826503discovered in
epididymis
hsa-miR-6724-5p54836504discovered in
epididymis
hsa-miR-675-3p54846505adrenocortical
tumor
hsa-miR-675-5p54856506adrenocortical
tumor
hsa-miR-676-3p54866507discovered in
female
reproductuve tract
hsa-miR-676-5p54876508discovered in
female
reproductuve tract
hsa-miR-708-3p54886509Various cancers
(lung, bladder,
pancreatic, ALL)
hsa-miR-708-5p54896510Various cancers
(lung, bladder,
pancreatic, ALL)
hsa-miR-71154906511cutaneous T-cell
lymphomas
hsa-miR-7-1-3p54916512Glioblast, brain,
prancreas
hsa-miR-71854926513blood
hsa-miR-7-2-3p54936514brain, pancreas
hsa-miR-744-3p54946515heart
hsa-miR-744-5p54956516embryonic stem
cells, heart
hsa-miR-758-3p54966517cholesterol
regulation and brain
hsa-miR-758-5p54976518cholesterol
regulation and brain
hsa-miR-75954986519
hsa-miR-7-5p54996520brain
hsa-miR-76055006521colonrectal and
breast cancer
hsa-miR-76155016522
hsa-miR-76255026523corneal epithelial
cells
hsa-miR-76455036524osteoblast
hsa-miR-76555046525rectal cancer
hsa-miR-766-3p55056526embryonic stem
cells
hsa-miR-766-5p55066527embryonic stem
cells
hsa-miR-767-3p55076528/
hsa-miR-767-5p55086529/
hsa-miR-769-3p55096530
hsa-miR-769-5p55106531
hsa-miR-770-5p55116532
hsa-miR-80255126533brain, epithelialdown symdrome
cells, hepatocytes
hsa-miR-873-3p55136534
hsa-miR-873-5p55146535
hsa-miR-87455156536cervical cancer,
lung cancer,
carcinoma
hsa-miR-875-3p55166537
hsa-miR-875-5p55176538
hsa-miR-876-3p55186539
hsa-miR-876-5p55196540
hsa-miR-877-3p55206541
hsa-miR-877-5p55216542
hsa-miR-885-3p55226543embryonic stem
cells
hsa-miR-885-5p55236544embryonic stem
cells
hsa-miR-88755246545
hsa-miR-888-3p55256546
hsa-miR-888-5p55266547
hsa-miR-88955276548
hsa-miR-89055286549epididymis
hsa-miR-891a55296550epididymisosteosarcoma
hsa-miR-891b55306551epididymis
hsa-miR-892a55316552epididymis
hsa-miR-892b55326553epididymis
hsa-miR-892c-3p55336554discovered in
epididymis
hsa-miR-892c-5p55346555discovered in
epididymis
hsa-miR-92055356556human testis
hsa-miR-92155366557human testismuscle invasive
bladder cancer
hsa-miR-92255376558human testis,multiple sclerosis,
neuronal tissuesAlcoholic liver
disease
hsa-miR-92455386559human testis
hsa-miR-92a-1-5p55396560endothelial cells
hsa-miR-92a-2-5p55406561endothelial cells
hsa-miR-92a-3p55416562endothelial cells,
CNS
hsa-miR-92b-3p55426563endothelial cells,
heart
hsa-miR-92b-5p55436564endothelial cells,
heart
hsa-miR-93355446565discovered in
cervical cancer
hsa-miR-93-3p55456566embryonic stembasal cell
cellscarcinoma
hsa-miR-93455466567discovered in
cervical cancer
hsa-miR-93555476568blood monoclearenergy
cellsmetabolism/
obesity,
medullablastoma/
neural stem cells
hsa-miR-93-5p55486569embryonic stem
cells
hsa-miR-93655496570skin
hsa-miR-937-3p55506571cervical cancer
hsa-miR-937-5p55516572cervical cancer
hsa-miR-93855526573Various cancer
cells
hsa-miR-939-3p55536574hepatocytes
hsa-miR-939-5p55546575hepatocytes
hsa-miR-9-3p55556576brainCancers and brain
diseases
hsa-miR-94055566577identified in
Cervical cancer
hsa-miR-94155576578Embryonic stem
cells
hsa-miR-94255586579lung cancer
hsa-miR-94355596580identified in
Cervical cancer
hsa-miR-94455606581various cancers
(cervical,
pancreatic,
colonrectal)
hsa-miR-9555616582various cancers
(pancreatic,
glioblastoma,
colorectal etc)
hsa-miR-9-5p55626583brainCancers and brain
disease
hsa-miR-96-3p55636584stem cellsvarious cancers
(prostate,
lymphoma, HCC,
etc) and
inflammation
hsa-miR-96-5p55646585stem cellsvarious cancers
(prostate,
lymphoma, HCC,
etc) and
inflammation
hsa-miR-98-3p55656586various cancerapoptosis
cells
hsa-miR-98-5p55666587various cancerapoptosis
cells
hsa-miR-99a-3p55676588hemapoietic cells
hsa-miR-99a-5p55686589hemapoietic cells
hsa-miR-99b-3p55696590hemapoietic cells,
embryonic stem
cells
hsa-miR-99b-5p55706591hemapoietic cells,
embryonic stem
cells

MicroRNAs that are enriched in specific types of immune cells are listed in Table 11. Furthermore, novel miroRNAs are discovered in the immune cells in the art through micro-array hybridization and microtome analysis (Jima D D et al, Blood, 2010, 116:e118-e127; Vaz C et al., BMC Genomics, 2010, 11,288, the content of each of which is incorporated herein by reference in its entirety). In Table 11, “HCC” represents hepatocellular carcinoma, “ALL” stands for acute lymphoblastic leukemia and “CLL” stands for chromic lymphocytic leukemia.

TABLE 11
microRNAs in immune cells
mirBStissues/cells
SEQSEQwithbiological
microRNAIDIDMicroRNAsassociated diseasesfunctions/targets
hsa-let-7a-2-3p25083529embryonic steminflammatory,tumor
cells, lung,various cancerssuppressor,
myeloid cells(lung, cervical,target to c-myc
breast, pancreatic,
etc)
hsa-let-7a-3p25093530embryonic steminflammatory,tumor
cell, lung,various cancerssuppressor,
myeloid cells(lung, cervical,target to c-myc
breast, pancreatic,
etc)
hsa-let-7a-5p25103531embryonic steminflammatory,tumor
cells, lung,various cancerssuppressor,
myeloid cells(lung, cervical,target to c-myc
breast, pancreatic,
etc)
hsa-let-7c25133534dendritic cellsvarious cacnerstumor
(cervical, pancreatic,suppressor
lung, esopphageal,apoptosis
etc)(target to BCL-
x1)
hsa-let-7e-3p25163537immune cellsvarious cancer cells,tumor
autoimmunitysuppressor
TLR signal pathway
in endotoxin
tolerance
hsa-let-7e-5p25173538immune cellsassociated withtumor
various cancer cellssuppressor
hsa-let-7f-1-3p25183539immune cells (Tassociated withtumor
cells)various cancer cellssuppressor
hsa-let-7f-2-3p25193540immune cells (Tassociated withtumor
cells)various cancer cellssuppressor
hsa-let-7f-5p25203541immune cells (Tassociated withtumor
cells)various cancer cellssuppressor
hsa-let-7g-3p25213542hematopoieticvarious cancer cellstumor
cells, adipose,(lung, breast, etc)suppressor
smooth muscle(target to
cellsNFkB, LOX1)
hsa-let-7g-5p25223543hematopoieticvarious cancer cellstumor
cells, adipose,(lung, breast, etc)suppressor
smooth muscle(target to
cellsNFkB, LOX1)
hsa-let-7i-3p25233544immune cellschronic lymphocytetumor
leukimiasuppressor
hsa-let-7i-5p25243545immune cellschronic lymphocytetumor
leukimiasuppressor
hsa-miR-10a-3p25303551hematopoeiticacute myeoidoncogene, cell
cellsleukemiagrowth
hsa-miR-10a-5p25413562hematopoieticacute myeloidoncogene, cell
cellsleukemiagrowth
hsa-miR-118425513572Hematopoieticdownregulated inpredited in the
cellsoral leukoplakiaintron 22 of F8
(OLK)gene
hsa-miR-125b-1-26163637hematopoieticvarious canceroncogene, cell
3pcells(ALL, prostate,differentiation
(monocytes),HCC, etc); TLR
brain (neuron)signal pathway in
endotoxin tolerance
hsa-miR-125b-2-26173638hematopoieticvarious canceroncogene cell
3pcells(ALL, prostate,differentiation
(monocytes),HCC etc); TLR
brain (neuron)signal pathway in
endotoxin tolerance
hsa-miR-125b-26183639hematopoieticvarious canceroncogene cell
5pcells, brain(Cutaneous T celldifferentiation
(neuron)lymphomas,
prostate, HCC, etc);
TLR signal pathway
in endotoxin
tolerance
hsa-miR-127926523673monocytes
hsa-miR-130a-3p26903711lung, monocytes,various cancerspro-angiogenic
vascular(basal cell
endothelial cellscarcinoma,
HCC, ovarian, etc),
drug resistance
hsa-miR-130a-5p26913712lung, monocytes,various cancerspro-angiogenic
vasscular(basal cell
endothelial cellscarcinoma,
HCC, ovarian, etc),
drug resistance
hsa-miR-132-3p26973718brain (neuron),
immune cells
hsa-miR-132-5p26993720brain (neuron),
immune cells
hsa-miR-142-3p27203741meyloid cells,tumor
hematopoiesis,suppressor,
APC cellsimmune
response
hsa-miR-142-5p27213742meyloid cells,immune
hematopoiesis,response
APC cells
hsa-miR-143-5p27233744vascular smoothincreased in serum
muscle, T-cellsafter virus infection
hsa-miR-146a-3p27303751immune cells,associated with
hematopoiesis, cartilage,CLL, TLR signal
pathway in
endotoxin tolerance
hsa-miR-146a-5p27313752immune cells,associated with
hematopoiesis,CLL, TLR signal
cartilage,pathway in
endotoxin tolerance
hsa-miR-146b-27323753immune cellscancers (thyroidimmune
3pcarcimona)response
hsa-miR-146b-27333754embryoid bodythyroid cancer,tumor invation,
5pcellsassociated with CLLmigration
hsa-miR-147a27363757Macrophageinflammatory
response
hsa-miR-147b27373758Macrophageinflammatory
response
hsa-miR-148a-3p27383759hematopoieticassociated with
cellsCLL, T-lineage
ALL
hsa-miR-148a-5p27393760hematopoieticassociated with
cellsCLL, T-lineage
ALL
hsa-miR-150-3p27443765hematopoiticcirculating plasma
cells (lymphoid)(acute myeloid
leukemia)
hsa-miR-150-5p27453766hematopoiticcirculating plasma
cells (lymphoid)(acute myeloid
leukemia)
hsa-miR-151b27483769immune cells (B-
cells)
hsa-miR-155-3p27563777T/B cells,associated with
monocytes, breastCLL, TLR signal
pathway in
endotoxin tolerance;
upregulated in B
cell lymphoma
(CLL) and other
cancers (breast,
lung, ovarian,
cervical, colorectal,
prostate)
hsa-miR-155-5p27573778T/B cells,associated with CLL,
monocytes, breastTLR signal
pathway in
endotoxin tolerance,
upregulated in B
cell lymphoma
(CLL) and other
cancers (breast,
lung, ovarian,
cervical, colorectal,
prostate)
hsa-miR-15a-3p27593780blood,chronic lymphocytic
lymphocyte,leukemia
hematopoietic
tissues (spleen)
hsa-miR-15a-5p27603781blood,chronic lymphocytic
lymphocyte,leukemia
hematopoietic
tissues (spleen)
hsa-miR-15b-3p27613782blood,cell cycle,
lymphocyte,proliferation
hematopoietic
tissues (spleen)
hsa-miR-15b-5p27623783blood,cell cycle,
lymphocyte,proliferation
hematopoietic
tissues (spleen)
hsa-miR-16-1-3p27633784embryonic stemchronic lymphocytic
cells, blood,leukemia
hematopoietic
tissues (spleen)
hsa-miR-16-2-3p27643785blood,
lymphocyte,
hematopoietic
tissues (spleen)
hsa-miR-16-5p27653786blood,
lymphocyte,
hematopoietic
tissues
hsa-miR-181a-3p27693790glioblast,
myeloid cells,
Embryonic stem
cells
hsa-miR-181a-5p27703791glioblast,
myeloid cells,
Embryonic stem
cells
hsa-miR-182-3p27763797immune cellscolonrectal cancer,immune
autoimmneresponse
hsa-miR-182-5p27783799lung, immuneautoimmuneimmune
cellsresponse
hsa-miR-197-3p28273848blood (myeloid),various cancers
other tissues(thyroid tumor,
leukemia, etc)
hsa-miR-197-5p28283849blood (myeloid),various cancers
other tissues(thyroid tumor,
leukemia, etc)
hsa-miR-21-3p28793099glioblast, Bloodautoimmune, heart
(meyloid cells),diseases, cancers
liver, vascular
endothelial cells
hsa-miR-214-3p28803901immune cells,varioua cancersimmune
pancreas(melanoma,response
pancreatic, ovarian)
hsa-miR-214-5p28813902immune cells,varioua cancersimmune
pancreas(melanoma,response
pancreatic, ovarian)
hsa-miR-21-5p28833904blood (myeloidautoimmune, heart
cells), liver,diseases, cancers
endothelial cells
hsa-miR-221-3p28943915endothelial cells,breastangiogenesis/vasculogenesis
immune cellscancer, upregulated
in thyroid cell
transformation
induced by
HMGA1, TLR
signal pathway in
endotoxin tolerance,
upregulated in T cell
ALL
hsa-miR-221-5p28953916endothelialbreastangiogenesis/vasculogenesis
cells, immunecancer, upregulated
cellsin thyroid cell
transformation
induced by
HMGA1, TLR
signal pathway in
endotoxin tolerance,
upregulated in T
cell ALL
hsa-miR-223-3p28983919meyloid cellsassociated with
CLL
hsa-miR-223-5p28993920meyloid cellsassociated with
CLL
hsa-miR-23b-3p29133934blood, myeloidcancers (renal
cellscancer,
glioblastoma,
prostate, etc)
and autoimmune
hsa-miR-23b-5p29143935blood, myeloidcancers (glioblastoma,
cellsprostate, etc) and
autoimmune
hsa-miR-24-1-5p29163937lung, myeloid
cells
hsa-miR-24-2-5p29173938lung, myeloid
cells
hsa-miR-24-3p29183939lung, myeloid
cells
hsa-miR-26a-1-29273948embryonic stemchronic lymphocytecell cycle and
3pcells, blood (Tleukemia and otherdifferentiation
cells)cancers
hsa-miR-26a-2-29283949blood (Tcells),chronic lymphocytecell cycle and
3pother tissuesleukemia and otherdifferentiation
cancers
hsa-miR-26a-5p29293950blood (Tcells),chronic lymphocytecell cycle and
other tissuesleukemia and otherdifferentiation
cancers
hsa-miR-26b-3p29303951hematopoietic
cells
hsa-miR-26b-5p29313952hematopoietic
cells
hsa-miR-27a-3p29323953myeloid cellsvarious cancer cells
hsa-miR-27a-5p29333954myeloid cellsvarious cancer cells
hsa-miR-27b-3p29343955myeloid cells,various cancer cellspro-angiogenic
vascular
endothelial cells
hsa-miR-28-3p29363957blood(immuneB/T cell lymphoma
cells)
hsa-miR-28-5p29373958blood(immuneB/T cell lymphoma
cells)
hsa-miR-290929393960T-Lymphocytes
hsa-miR-29a-3p29483969immuno system,various cancers,tumor
colonrectunneurodegenativesuppression,
diseaseimmune
modulation
(mir-29 family)
hsa-miR-29a-5p29493970immuno system,various cancers,adaptive
colonrectunneurodegenativeimmunity
disease
hsa-miR-29b-1-29503971immuno systemassociated withadaptive
5pCLL, other cancers,immunity
neurodegenative
disease
hsa-miR-29b-2-29513972immuno systemassociated withadaptive
5pCLL, other cancers,immunity
hsa-miR-29b-3p29523973immuno systemassociated withadaptive
CLL, other cancersimmunity
hsa-miR-29c-3p29533974immuno systemassociated withadaptive
CLL, other cancersimmunity
hsa-miR-29c-5p29543975immuno systemassociated withadaptive
CLL, other cancersimmunity
hsa-miR-30e-3p29844005myeloid cells,
glia cells
hsa-miR-30e-5p29854006myeloid cells,
glia cells
hsa-miR-331-5p31304151lymphocytes
hsa-miR-339-3p31374158immune cells
hsa-miR-339-5p31384159immune cells
hsa-miR-345-3p31474168hematopoieticincreased in
cellsfollicular
lymphoma (53),
other cancers
hsa-miR-345-5p31484169hematopoieticincreased in
cellsfollicular
lymphoma (53)
hsa-miR-34631494170immume cellscancers and
autoimmune
hsa-miR-34a-3p31504171breast, myeloidgastric cancer,tumor
cells, ciliatedCLL, othersuppressor, p53
epithelial cellsinducible
hsa-miR-34a-5p31514172breast, myeloidgastric cancer,tumor
cells, ciliatedCLL, othersuppressor, p53
epithelial cellsinducible
hsa-miR-363-3p31934214kidney stem cell,
blood cells
hsa-miR-363-5p31944215kidney stem cell,
blood cells
hsa-miR-37232774298hematopoietic
cells, lung,
placental (blood)
hsa-miR-377-3p32944315hematopoietic
cells
hsa-miR-377-5p32954316hematopoietic
cells
hsa-miR-493-3p49475968myeloid cells,
pancreas (islet)
hsa-miR-493-5p49485969myeloid cells,
pancreas (islet)
hsa-miR-542-3p51066127monocytestargets to
survivin,
introduce
growth arrest
hsa-miR-548b-51576178immune cells
5pfrontal cortex
hsa-miR-548c-5p51596180immune cells
frontal cortex
hsa-miR-548i51686189embryonic stem
cells (41),
immune cells
hsa-miR-548j51696190immune cells
hsa-miR-548n51736194embryonic stem
cells, immune
cells
hsa-miR-574-3p52796300blood (myeloidincreased in
cells)follicular
lymphoma (53)
hsa-miR-59853106331in blood
lymphocytes
(PBL)
hsa-miR-93555476568identified inassociated with
human cervicalenergy
cancermetabolism/obesity,
bloodmedullablastoma/neural
mononuclearstem cells
cells
hsa-miR-99a-3p55676588hemapoietic cells
hsa-miR-99a-5p55686589hemapoietic
cells, plasma
(exosome)
hsa-miR-99b-3p55696590hemapoietic
cells, Embryonic
stem cells,
hsa-miR-99b-5p55706591hemapoietic
cells, Embryonic
stem cells,
plasma (exosome)

Herein, in a signal-sensor polynucleotide (such as a primary construct or a mRNA molecule), the terms “modification” or, as appropriate, “modified” refer to modification with respect to A, G, U or C ribonucleotides. Generally, herein, these terms are not intended to refer to the ribonucleotide modifications in naturally occurring 5′-terminal mRNA cap moieties. In a polypeptide, the term “modification” refers to a modification as compared to the canonical set of 20 amino acids.

The modifications may be various distinct modifications. In some embodiments, the coding region, the flanking regions and/or the terminal regions may contain one, two, or more (optionally different) nucleoside or nucleotide modifications. In some embodiments, a modified signal-sensor polynucleotide, primary construct, or mmRNA introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified signal-sensor polynucleotide, primary construct, or mmRNA.

The signal-sensor polynucleotides, primary constructs, and mmRNA can include any useful modification, such as to the sugar, the nucleobase, or the internucleoside linkage (e.g. to a linking phosphate/to a phosphodiester linkage/to the phosphodiester backbone). One or more atoms of a pyrimidine nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro). In certain embodiments, modifications (e.g., one or more modifications) are present in each of the sugar and the internucleoside linkage. Modifications according to the present invention may be modifications of ribonucleic acids (RNAs) to deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs) or hybrids thereof). Additional modifications are described herein.

As described herein, in some embodiments, the signal-sensor polynucleotides, primary constructs, and mmRNA of the invention do not substantially induce an innate immune response of a cell into which the mRNA is introduced. Features of an induced innate immune response include 1) increased expression of pro-inflammatory cytokines, 2) activation of intracellular PRRs (RIG-I, MDA5, etc, and/or 3) termination or reduction in protein translation. In other embodiments, an immune response is induced.

In certain embodiments, it may desirable to intracellularly degrade a modified nucleic acid molecule introduced into the cell. For example, degradation of a modified nucleic acid molecule may be preferable if precise timing of protein production is desired. Thus, in some embodiments, the invention provides a modified nucleic acid molecule containing a degradation domain, which is capable of being acted on in a directed manner within a cell.

In another aspect, the present disclosure provides signal-sensor polynucleotides comprising a nucleoside or nucleotide that can disrupt the binding of a major groove interacting, e.g. binding, partner with the polynucleotide (e.g., where the modified nucleotide has decreased binding affinity to major groove interacting partner, as compared to an unmodified nucleotide).

The signal-sensor polynucleotides, primary constructs, and mmRNA can optionally include other agents (e.g., RNAi-inducing agents, RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes, catalytic DNA, tRNA, RNAs that induce triple helix formation, aptamers, vectors, etc.). In some embodiments, the signal-sensor polynucleotides, primary constructs, or mmRNA may include one or more messenger RNAs (mRNAs) and one or more modified nucleoside or nucleotides (e.g., mmRNA molecules). Details for these signal-sensor polynucleotides, primary constructs, and mmRNA follow.

Signal-Sensor Polynucleotides and Primary Constructs

The signal-sensor polynucleotides, primary constructs, and mmRNA of the invention includes a first region of linked nucleosides encoding an oncology-related polypeptide of interest, a first flanking region located at the 5′ terminus of the first region, and a second flanking region located at the 3′ terminus of the first region.

In some embodiments, the signal-sensor polynucleotide, primary construct, or mmRNA are constructed according to the methods and modifications of International Application PCT/US12/058519 filed Oct. 3, 2012 (M9), the contents of which are incorporated herein by reference in their entirety.

The signal-sensor polynucleotides, primary constructs, and mmRNA can optionally include 5′ and/or 3′ flanking regions, which are described herein.

Signal-Sensor Modified RNA (mmRNA) Molecules

The present invention also includes the building blocks, e.g., modified ribonucleosides, modified ribonucleotides, of modified signal-sensor mRNA (mmRNA) molecules. For example, these building blocks can be useful for preparing the signal-sensor polynucleotides, primary constructs, or mmRNA of the invention. Such building blocks are taught in co-pending International Application PCT/US12/058519 filed Oct. 3, 2012 (M9), the contents of which are incorporated herein by reference in their entirety.

Modifications on the Nucleobase

The present disclosure provides for modified nucleosides and nucleotides. As described herein “nucleoside” is defined as a compound containing a sugar molecule (e.g., a pentose or ribose) or a derivative thereof in combination with an organic base (e.g., a purine or pyrimidine) or a derivative thereof (also referred to herein as “nucleobase”). As described herein, “nucleotide” is defined as a nucleoside including a phosphate group. In some embodiments, the nucleosides and nucleotides described herein are generally chemically modified on the major groove face. Exemplary non-limiting modifications include an amino group, a thiol group, an alkyl group, a halo group, or any described herein. The modified nucleotides may by synthesized by any useful method, as described herein (e.g., chemically, enzymatically, or recombinantly to include one or more modified or non-natural nucleosides).

The modified nucleosides and nucleotides can include a modified nucleobase. Examples of nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine, and uracil. Examples of nucleobase found in DNA include, but are not limited to, adenine, guanine, cytosine, and thymine. These nucleobases can be modified or wholly replaced to provide signal-sensor polynucleotides, primary constructs, or mmRNA molecules having enhanced properties. For example, the nucleosides and nucleotides described herein can be chemically modified. In some embodiments, chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.

Modifications on the Internucleoside Linkage

The modified nucleotides, which may be incorporated into a signal-sensor polynucleotide, primary construct, or mmRNA molecule, can be modified on the internucleoside linkage (e.g., phosphate backbone). Herein, in the context of the polynucleotide backbone, the phrases “phosphate” and “phosphodiester” are used interchangeably. Backbone phosphate groups can be modified by replacing one or more of the oxygen atoms with a different substituent. Further, the modified nucleosides and nucleotides can include the wholesale replacement of an unmodified phosphate moiety with another internucleoside linkage as described herein. Examples of modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, boranophosphates, boranophosphate esters, hydrogen phosphonates, phosphoramidates, phosphorodiamidates, alkyl or aryl phosphonates, and phosphotriesters. Phosphorodithioates have both non-linking oxygens replaced by sulfur. The phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoramidates), sulfur (bridged phosphorothioates), and carbon (bridged methylene-phosphonates).

The α-thio substituted phosphate moiety is provided to confer stability to RNA and DNA polymers through the unnatural phosphorothioate backbone linkages. Phosphorothioate DNA and RNA have increased nuclease resistance and subsequently a longer half-life in a cellular environment. Phosphorothioate linked signal-sensor polynucleotides, primary constructs, or mmRNA molecules are expected to also reduce the innate immune response through weaker binding/activation of cellular innate immune molecules.

In specific embodiments, a modified nucleoside includes an alpha-thio-nucleoside (e.g., 5′-O-(1-thiophosphate)-adenosine, 5′-O-(1-thiophosphate)-cytidine (α-thio-cytidine), 5′-O-(1-thiophosphate)-guanosine, 5′-O-(1-thiophosphate)-uridine, or 5′-O-(1-thiophosphate)-pseudouridine).

Other internucleoside linkages that may be employed according to the present invention, including internucleoside linkages which do not contain a phosphorous atom, are described herein below.

Combinations of Modified Sugars, Nucleobases, and Internucleoside Linkages

The signal-sensor polynucleotides, primary constructs, and mmRNA of the invention can include a combination of modifications to the sugar, the nucleobase, and/or the internucleoside linkage. These combinations can include any one or more modifications described herein or in International Application PCT/US12/058519 filed Oct. 3, 2012 (M9), the contents of which are incorporated herein by reference in their entirety.

Synthesis of Signal-Sensor Primary Constructs, and mmRNA Molecules

The signal-sensor polypeptides, primary constructs, and mmRNA molecules for use in accordance with the invention may be prepared according to any useful technique, as described herein. The modified nucleosides and nucleotides used in the synthesis of signal-sensor polynucleotides, primary constructs, and mmRNA molecules disclosed herein can be prepared from readily available starting materials using the following general methods and procedures. Where typical or preferred process conditions (e.g., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are provided, a skilled artisan would be able to optimize and develop additional process conditions. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.

The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C) infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.

Preparation of signal-sensor polynucleotides, primary constructs, and mRNA molecules of the present invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991, which is incorporated herein by reference in its entirety.

The reactions of the processes described herein can be carried out in suitable solvents, which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.

Resolution of racemic mixtures of modified nucleosides and nucleotides (e.g., mmRNA molecules) can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallization using a “chiral resolving acid” which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids. Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.

Modified nucleosides and nucleotides (e.g., building block molecules) can be prepared according to the synthetic methods described in Ogata et al., J. Org. Chem. 74:2585-2588 (2009); Purmal et al., Nucl. Acids Res. 22(1): 72-78, (1994); f*ckuhara et al., Biochemistry, 1(4): 563-568 (1962); and Xu et al., Tetrahedron, 48(9): 1729-1740 (1992), each of which are incorporated by reference in their entirety.

The signal-sensor polynucleotides, primary constructs, and mmRNA of the invention may or may not be uniformly modified along the entire length of the molecule. For example, one or more or all types of nucleotide (e.g., purine or pyrimidine, or any one or more or all of A, G, U, C) may or may not be uniformly modified in a polynucleotide of the invention, or in a given predetermined sequence region thereof (e.g. one or more of the sequence regions represented in FIG. 1). In some embodiments, all nucleotides X in a signal-sensor polynucleotide of the invention (or in a given sequence region thereof) are modified, wherein X may any one of nucleotides A, G, U, C, or any one of the combinations A+G, A+U, A+C, G+U, G+C, U+C, A+G+U, A+G+C, G+U+C or A+G+C.

Different sugar modifications, nucleotide modifications, and/or internucleoside linkages (e.g., backbone structures) may exist at various positions in the signal-sensor polynucleotide, primary construct, or mmRNA. One of ordinary skill in the art will appreciate that the nucleotide analogs or other modification(s) may be located at any position(s) of a signal-sensor polynucleotide, primary construct, or mmRNA such that the function of the signal-sensor polynucleotide, primary construct, or mmRNA is not substantially decreased. A modification may also be a 5′ or 3′ terminal modification. The signal-sensor polynucleotide, primary construct, or mmRNA may contain from about 1% to about 100% modified nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e. any one or more of A, G, U or C) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100%).

In some embodiments, the signal-sensor polynucleotide, primary construct, or mmRNA includes a modified pyrimidine (e.g., a modified uracil/uridine/U or modified cytosine/cytidine/C). In some embodiments, the uracil or uridine (generally: U) in the signal-sensor polynucleotide, primary construct, or mmRNA molecule may be replaced with from about 1% to about 100% of a modified uracil or modified uridine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified uracil or modified uridine). The modified uracil or uridine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein). In some embodiments, the cytosine or cytidine (generally: C) in the signal-sensor polynucleotide, primary construct, or mmRNA molecule may be replaced with from about 1% to about 100% of a modified cytosine or modified cytidine (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%, from 70% to 90%, from 70% to 95%, from 70% to 100%, from 80% to 90%, from 80% to 95%, from 80% to 100%, from 90% to 95%, from 90% to 100%, and from 95% to 100% of a modified cytosine or modified cytidine). The modified cytosine or cytidine can be replaced by a compound having a single unique structure or by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures, as described herein).

Combinations of Nucleotides

Further examples of modified nucleotides and modified nucleotide combinations are provided in International Application PCT/US12/058519 filed Oct. 3, 2012 (M9) the contents of which are incorporated herein by reference in their entirety.

In some embodiments, at least 25% of the cytidines are replaced (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).

In some embodiments, at least 25% of the uracils are replaced (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).

In some embodiments, at least 25% of the cytidines are replaced, and at least 25% of the uracils are replaced (e.g., at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%).

Formulation, Administration, Delivery and Dosing

The present invention provides signal-sensor polynucleotides, primary constructs and mmRNA compositions and complexes in combination with one or more pharmaceutically acceptable excipients. Pharmaceutical compositions may optionally comprise one or more additional active substances, e.g. therapeutically and/or prophylactically active substances. General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference).

In some embodiments, compositions are administered to humans, human patients or subjects. For the purposes of the present disclosure, the phrase “active ingredient” generally refers to signal-sensor polynucleotides, primary constructs and mmRNA to be delivered as described herein.

Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.

Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing the active ingredient into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging the product into a desired single- or multi-dose unit.

A pharmaceutical composition in accordance with the invention may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.

Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1% and 100%, e.g., between 0.5 and 50%, between 1-30%, between 5-80%, at least 80% (w/w) active ingredient.

Formulations

The signal-sensor polynucleotide, primary construct, and mmRNA of the invention can be formulated using one or more excipients to: (1) increase stability; (2) increase cell transfection; (3) permit the sustained or delayed release (e.g., from a depot formulation of the signal-sensor polynucleotide, primary construct, or mmRNA); (4) alter the biodistribution (e.g., target the polynucleotide, primary construct, or mmRNA to specific tissues or cell types); (5) increase the translation of encoded protein in vivo; and/or (6) alter the release profile of encoded protein in vivo. In addition to traditional excipients such as any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, excipients of the present invention can include, without limitation, lipidoids, liposomes, lipid nanoparticles, polymers, lipoplexes, core-shell nanoparticles, peptides, proteins, cells transfected with signal-sensor polynucleotide, primary construct, or mmRNA (e.g., for transplantation into a subject), hyaluronidase, nanoparticle mimics and combinations thereof. Further, the signal-sensor polynucleotide, primary construct, or mmRNA of the present invention may be formulated using self-assembled nucleic acid nanoparticles.

Accordingly, the formulations of the invention can include one or more excipients, each in an amount that together increases the stability of the signal-sensor polynucleotide, primary construct, or mmRNA, increases cell transfection by the signal-sensor polynucleotide, primary construct, or mmRNA, increases the expression of polynucleotide, primary construct, or mmRNA encoded protein, and/or alters the release profile of signal-sensor polynucleotide, primary construct, or mmRNA encoded proteins. Further, the primary construct and mmRNA of the present invention may be formulated using self-assembled nucleic acid nanoparticles.

Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of associating the active ingredient with an excipient and/or one or more other accessory ingredients.

A pharmaceutical composition in accordance with the present disclosure may be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. As used herein, a “unit dose” refers to a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient may generally be equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage including, but not limited to, one-half or one-third of such a dosage.

Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject being treated and further depending upon the route by which the composition is to be administered. For example, the composition may comprise between 0.1% and 99% (w/w) of the active ingredient.

In some embodiments, the formulations described herein may contain at least one signal-sensor mmRNA. As a non-limiting example, the formulations may contain 1, 2, 3, 4 or 5 signal-sensor mmRNA. In one embodiment the formulation may contain modified mRNA encoding proteins selected from categories such as, proteins. In one embodiment, the formulation contains at least three signal-sensor modified mRNA encoding oncology-related proteins. In one embodiment, the formulation contains at least five signal-sensor modified mRNA encoding oncology-related proteins.

Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes, but is not limited to, any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, and the like, as suited to the particular dosage form desired. Various excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, Md., 2006; incorporated herein by reference). The use of a conventional excipient medium may be contemplated within the scope of the present disclosure, except insofar as any conventional excipient medium may be incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition.

In some embodiments, the particle size of the lipid nanoparticle may be increased and/or decreased. The change in particle size may be able to help counter biological reaction such as, but not limited to, inflammation or may increase the biological effect of the signal-sensor modified mRNA delivered to mammals.

Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, surface active agents and/or emulsifiers, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in the pharmaceutical formulations of the invention.

Pharmaceutical compositions of the present invention may comprise at least one adjuvant which may be a chemo-adjuvant. Non-limiting examples of chemo-adjuvants and delivery systems which comprises a chemo-adjuvant are described in International Patent Publication No. WO2013134349, the contents of which is herein incorporated by reference in its entirety. The chemo-adjuvant may be bonded to, non-covalently bonded to or encapsulated within a delivery vehicle described herein.

Lipidoids

The synthesis of lipidoids has been extensively described and formulations containing these compounds are particularly suited for delivery of signal-sensor polynucleotides, primary constructs or mmRNA (see Mahon et al., Bioconjug Chem. 2010 21:1448-1454; Schroeder et al., J Intern Med. 2010 267:9-21; Akinc et al., Nat Biotechnol. 2008 26:561-569; Love et al., Proc Natl Acad Sci USA. 2010 107:1864-1869; Siegwart et al., Proc Natl Acad Sci USA. 2011 108:12996-3001; all of which are incorporated herein in their entireties).

While these lipidoids have been used to effectively deliver double stranded small interfering RNA molecules in rodents and non-human primates (see Akinc et al., Nat Biotechnol. 2008 26:561-569; Frank-Kamenetsky et al., Proc Natl Acad Sci USA. 2008 105:11915-11920; Akinc et al., Mol Ther. 2009 17:872-879; Love et al., Proc Natl Acad Sci USA. 2010 107:1864-1869; Leuschner et al., Nat Biotechnol. 2011 29:1005-1010; all of which is incorporated herein in their entirety), the present disclosure describes their formulation and use in delivering single stranded signal-sensor polynucleotides, primary constructs, or mmRNA. Complexes, micelles, liposomes or particles can be prepared containing these lipidoids and therefore, can result in an effective delivery of the signal-sensor polynucleotide, primary construct, or mmRNA, as judged by the production of an encoded protein, following the injection of a lipidoid formulation via localized and/or systemic routes of administration. Lipidoid complexes of signal-sensor polynucleotides, primary constructs, or mmRNA can be administered by various means including, but not limited to, intravenous, intramuscular, or subcutaneous routes.

In vivo delivery of nucleic acids may be affected by many parameters, including, but not limited to, the formulation composition, nature of particle PEGylation, degree of loading, oligonucleotide to lipid ratio, and biophysical parameters such as particle size (Akinc et al., Mol Ther. 2009 17:872-879; herein incorporated by reference in its entirety). As an example, small changes in the anchor chain length of poly(ethylene glycol) (PEG) lipids may result in significant effects on in vivo efficacy. Formulations with the different lipidoids, including, but not limited to penta[3-(1-laurylaminopropionyl)]-triethylenetetramine hydrochloride (TETA-5LAP; aka 98N12-5, see Murugaiah et al., Analytical Biochemistry, 401:61 (2010)), C12-200 (including derivatives and variants), and MD1, can be tested for in vivo activity.

The lipidoid referred to herein as “98N12-5” is disclosed by Akinc et al., Mol Ther. 2009 17:872-879 and is incorporated by reference in its entirety.

The lipidoid referred to herein as “C12-200” is disclosed by Love et al., Proc Natl Acad Sci USA. 2010 107:1864-1869 and Liu and Huang, Molecular Therapy. 2010 669-670; both of which are herein incorporated by reference in their entirety. The lipidoid formulations can include particles comprising either 3 or 4 or more components in addition to signal-sensor polynucleotide, primary construct, or mmRNA. As an example, formulations with certain lipidoids, include, but are not limited to, 98N12-5 and may contain 42% lipidoid, 48% cholesterol and 10% PEG (C14 alkyl chain length). As another example, formulations with certain lipidoids, include, but are not limited to, C12-200 and may contain 50% lipidoid, 10% disteroylphosphatidyl choline, 38.5% cholesterol, and 1.5% PEG-DMG.

Combinations of different lipidoids may be used to improve the efficacy of signal-sensor polynucleotide, primary construct, or mmRNA directed protein production as the lipidoids may be able to increase cell transfection by the signal-sensor polynucleotide, primary construct, or mmRNA; and/or increase the translation of encoded oncology-related protein (see Whitehead et al., Mol. Ther. 2011, 19:1688-1694, herein incorporated by reference in its entirety).

In some embodiments, the particle size of the lipid nanoparticle may be increased and/or decreased. The change in particle size may be able to help counter biological reaction such as, but not limited to, inflammation or may increase the biological effect of, the signal-sensor polynucleotide, primary construct, or mmRNA delivered to subjects.

Liposomes, Lipoplexes, and Lipid Nanoparticles

The signal-sensor polynucleotide, primary construct, and mmRNA of the invention can be formulated using one or more liposomes, lipoplexes, or lipid nanoparticles. In one embodiment, pharmaceutical compositions of signal-sensor polynucleotide, primary construct, or mmRNA include liposomes. Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as a delivery vehicle for the administration of nutrients and pharmaceutical formulations. Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 and 500 nm in diameter. Liposome design may include, but is not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis. Liposomes may contain a low or a high pH in order to improve the delivery of the pharmaceutical formulations.

The formation of liposomes may depend on the physicochemical characteristics such as, but not limited to, the pharmaceutical formulation entrapped and the liposomal ingredients, the nature of the medium in which the lipid vesicles are dispersed, the effective concentration of the entrapped substance and its potential toxicity, any additional processes involved during the application and/or delivery of the vesicles, the optimization size, polydispersity and the shelf-life of the vesicles for the intended application, and the batch-to-batch reproducibility and possibility of large-scale production of safe and efficient liposomal products.

In one embodiment, pharmaceutical compositions described herein may include, without limitation, liposomes such as those formed from 1,2-dioleyloxy-N,N-dimethylaminopropane (DODMA) liposomes, DiLa2 liposomes from Marina Biotech (Bothell, Wash.), 1,2-dilinoleyloxy-3-dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4-(2-dimethylaminoethyl)-[1,3]-dioxolane (DLin-KC2-DMA), and MC3 (US20100324120; herein incorporated by reference in its entirety) and liposomes which may deliver small molecule drugs such as, but not limited to, DOXIL® from Janssen Biotech, Inc. (Horsham, Pa.).

In one embodiment, pharmaceutical compositions described herein may include, without limitation, liposomes such as those formed from the synthesis of stabilized plasmid-lipid particles (SPLP) or stabilized nucleic acid lipid particle (SNALP) that have been previously described and shown to be suitable for oligonucleotide delivery in vitro and in vivo (see Wheeler et al. Gene Therapy. 1999 6:271-281; Zhang et al. Gene Therapy. 1999 6:1438-1447; Jeffs et al. Pharm Res. 2005 22:362-372; Morrissey et al., Nat Biotechnol. 2005 2:1002-1007; Zimmermann et al., Nature. 2006 441:111-114; Heyes et al. J Contr Rel. 2005 107:276-287; Semple et al. Nature Biotech. 2010 28:172-176; Judge et al. J Clin Invest. 2009 119:661-673; deFougerolles Hum Gene Ther. 2008 19:125-132; all of which are incorporated herein in their entireties.) The original manufacture method by Wheeler et al. was a detergent dialysis method, which was later improved by Jeffs et al. and is referred to as the spontaneous vesicle formation method. The liposome formulations are composed of 3 to 4 lipid components in addition to the signal-sensor polynucleotide, primary construct, or mmRNA. As an example a liposome can contain, but is not limited to, 55% cholesterol, 20% disteroylphosphatidyl choline (DSPC), 10% PEG-S-DSG, and 15% 1,2-dioleyloxy-N,N-dimethylaminopropane (DODMA), as described by Jeffs et al. As another example, certain liposome formulations may contain, but are not limited to, 48% cholesterol, 20% DSPC, 2% PEG-c-DMA, and 30% cationic lipid, where the cationic lipid can be 1,2-distearloxy-N,N-dimethylaminopropane (DSDMA), DODMA, DLin-DMA, or 1,2-dilinolenyloxy-3-dimethylaminopropane (DLenDMA), as described by Heyes et al.

In one embodiment, pharmaceutical compostions may include liposomes which may be formed to deliver signal-sensor mmRNA which may encode at least one immunogen. The mmRNA may be encapsulated by the liposome and/or it may be contained in an aqueous core which may then be encapsulated by the liposome (see International Pub. Nos. WO2012031046, WO2012031043, WO201203091 and WO2012006378 herein incorporated by reference in their entireties). In another embodiment, the signal-sensor mmRNA which may encode an immunogen may be formulated in a cationic oil-in-water emulation where the emulsion particle comprises an oil core and a cationic lipid which can interact with the signal-sensor mmRNA anchoring the molecule to the emulsion particle (see International Pub. No. WO2012006380). In yet another embodiment, the lipid formulation may include at least cationic lipid, a lipid which may enhance transfection and a least one lipid which contains a hydrophilic head group linked to a lipid moiety (International Pub. No. WO2011076807 and U.S. Pub. No. 20110200582; herein incorporated by reference in their entireties). In another embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA encoding an immunogen may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers (see U.S. Pub. No. 20120177724, herein incorporated by reference in its entirety).

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be formulated in a lipid vesicle which may have crosslinks between functionalized lipid bilayers.

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be formulated in a lipid-polycation complex. The formation of the lipid-polycation complex may be accomplished by methods known in the art and/or as described in U.S. Pub. No. 20120178702, herein incorporated by reference in its entirety. As a non-limiting example, the polycation may include a cationic peptide or a polypeptide such as, but not limited to, polylysine, polyornithine and/or polyarginine. In another embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be formulated in a lipid-polycation complex which may further include a neutral lipid such as, but not limited to, cholesterol or dioleoyl phosphatidylethanolamine (DOPE).

The liposome formulation may be influenced by, but not limited to, the selection of the cationic lipid component, the degree of cationic lipid saturation, the nature of the PEGylation, ratio of all components and biophysical parameters such as size. In one example by Semple et al. (Semple et al. Nature Biotech. 2010 28:172-176), the liposome formulation was composed of 57.1% cationic lipid, 7.1% dipalmitoylphosphatidylcholine, 34.3% cholesterol, and 1.4% PEG-c-DMA. As another example, changing the composition of the cationic lipid could more effectively deliver siRNA to various antigen presenting cells (Basha et al. Mol Ther. 2011 19:2186-2200; herein incorporated by reference in its entirety).

In some embodiments, the ratio of PEG in the LNP formulations may be increased or decreased and/or the carbon chain length of the PEG lipid may be modified from C14 to C18 to alter the pharmaco*kinetics and/or biodistribution of the LNP formulations. As a non-limiting example, LNP formulations may contain 1-5% of the lipid molar ratio of PEG-c-DOMG as compared to the cationic lipid, DSPC and cholesterol. In another embodiment the PEG-c-DOMG may be replaced with a PEG lipid such as, but not limited to, PEG-DSG (1,2-Distearoyl-sn-glycerol, methoxypolyethylene glycol) or PEG-DPG (1,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol). The cationic lipid may be selected from any lipid known in the art such as, but not limited to, DLin-MC3-DMA, DLin-DMA, C12-200 and DLin-KC2-DMA.

In one embodiment, the LNP formulations of the signal-sensor polynucleotides, primary constructs and/or mmRNA may contain PEG-c-DOMG 3% lipid molar ratio. In another embodiment, the LNP formulations of the signal-sensor polynucleotides, primary constructs and/or mmRNA may contain PEG-c-DOMG 1.5% lipid molar ratio.

In one embodiment, the pharmaceutical compositions of the signal-sensor polynucleotides, primary constructs and/or mmRNA may include at least one of the PEGylated lipids described in International Publication No. 2012099755, herein incorporated by reference.

In one embodiment, the pharmaceutical compositions may be formulated in liposomes such as, but not limited to, DiLa2 liposomes (Marina Biotech, Bothell, Wash.), SMARTICLES® (Marina Biotech, Bothell, Wash.), neutral DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine) based liposomes (e.g., siRNA delivery for ovarian cancer (Landen et al. Cancer Biology & Therapy 2006 5(12)1708-1713)) and hyaluronan-coated liposomes (Quiet Therapeutics, Israel).

In some embodiments the liposome may be a liposomal nanostructure which has been formulated for treatment of cancers and other diseases or to control the cholesterol metabolism in cells. The liposome nanostructure may also comprise a scavenger receptor type B-1 (SR-B1) in order to kill cancer cells. Non-limiting examples of liposomal nanostructures, which may be used with the signal-sensor polynucleotides described herein, are described in International Publication No. WO2013126776, the contents of which are herein incorporated by reference in its entirety.

In one embodiment, the liposomes described herein may comprise at least one immunomodulator such as, but not limited to, cytokines. Formulations and methods of using the liposomes comprising at least one immunomodulator are described in International Publication No WO2013129935 and WO2013129936, the contents of each of which are herein incorporated by reference in their entirety. As a non-limiting example, the liposomes comprising at least one immunomodulator may be used in the treatment of cancer. The liposomes comprising an immunomodulator may comprise a signal-sensor polynucleotide described herein. As a non-limiting example, the liposome comprising an immunomodulator may be used in a combination with at least one antibody such as the particulate or vesicular immunomodulators described in International Publication No WO2013129936, the contents of which are herein incorporated by reference in its entirety.

Lipid nanoparticle formulations may be improved by replacing the cationic lipid with a biodegradable cationic lipid which is known as a rapidly eliminated lipid nanoparticle (reLNP). Ionizable cationic lipids, such as, but not limited to, DLinDMA, DLin-KC2-DMA, and DLin-MC3-DMA, have been shown to accumulate in plasma and tissues over time and may be a potential source of toxicity. The rapid metabolism of the rapidly eliminated lipids can improve the tolerability and therapeutic index of the lipid nanoparticles by an order of magnitude from a 1 mg/kg dose to a 10 mg/kg dose in rat. Inclusion of an enzymatically degraded ester linkage can improve the degradation and metabolism profile of the cationic component, while still maintaining the activity of the reLNP formulation. The ester linkage can be internally located within the lipid chain or it may be terminally located at the terminal end of the lipid chain. The internal ester linkage may replace any carbon in the lipid chain.

In one embodiment, the internal ester linkage may be located on either side of the saturated carbon.

In one embodiment, an immune response may be elicited by delivering a lipid nanoparticle which may include a nanospecies, a polymer and an immunogen. (U.S. Publication No. 20120189700 and International Publication No. WO2012099805; herein incorporated by reference in their entireties). The polymer may encapsulate the nanospecies or partially encapsulate the nanospecies. The immunogen may be a recombinant oncology-related protein, a signal-sensor modified RNA and/or a primary construct described herein. In one embodiment, the lipid nanoparticle may be formulated for use in a vaccine such as, but not limited to, against a pathogen.

Lipid nanoparticles may be engineered to alter the surface properties of particles so the lipid nanoparticles may penetrate the mucosal barrier. Mucus is located on mucosal tissue such as, but not limited to, oral (e.g., the buccal and esophageal membranes and tonsil tissue), ophthalmic, gastrointestinal (e.g., stomach, small intestine, large intestine, colon, rectum), nasal, respiratory (e.g., nasal, pharyngeal, tracheal and bronchial membranes), genital (e.g., vagin*l, cervical and urethral membranes). Nanoparticles larger than 10-200 nm which are preferred for higher drug encapsulation efficiency and the ability to provide the sustained delivery of a wide array of drugs have been thought to be too large to rapidly diffuse through mucosal barriers. Mucus is continuously secreted, shed, discarded or digested and recycled so most of the trapped particles may be removed from the mucosal tissue within seconds or within a few hours. Large polymeric nanoparticles (200 nm-500 nm in diameter) which have been coated densely with a low molecular weight polyethylene glycol (PEG) diffused through mucus only 4 to 6-fold lower than the same particles diffusing in water (Lai et al. PNAS 2007 104(5):1482-487; Lai et al. Adv Drug Deliv Rev. 2009 61(2): 158-171; herein incorporated by reference in their entirety). The transport of nanoparticles may be determined using rates of permeation and/or fluorescent microscopy techniques including, but not limited to, fluorescence recovery after photobleaching (FRAP) and high resolution multiple particle tracking (MPT).

The lipid nanoparticle engineered to penetrate mucus may comprise a polymeric material (i.e. a polymeric core) and/or a polymer-vitamin conjugate and/or a tri-block co-polymer. The polymeric material may include, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, poly(styrenes), polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyeneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates. The polymeric material may be biodegradable and/or biocompatible. Non-limiting examples of specific polymers include poly(caprolactone) (PCL), ethylene vinyl acetate polymer (EVA), poly(lactic acid) (PLA), poly(L-lactic acid) (PLLA), poly(glycolic acid) (PGA), poly(lactic acid-co-glycolic acid) (PLGA), poly(L-lactic acid-co-glycolic acid) (PLLGA), poly(D,L-lactide) (PDLA), poly(L-lactide) (PLLA), poly(D,L-lactide-co-caprolactone), poly(D,L-lactide-co-caprolactone-co-glycolide), poly(D,L-lactide-co-PEO-co-D,L-lactide), poly(D,L-lactide-co-PPO-co-D,L-lactide), polyalkyl cyanoacrylate, polyurethane, poly-L-lysine (PLL), hydroxypropyl methacrylate (HPMA), polyethyleneglycol, poly-L-glutamic acid, poly(hydroxy acids), polyanhydrides, polyorthoesters, poly(ester amides), polyamides, poly(ester ethers), polycarbonates, polyalkylenes such as polyethylene and polypropylene, polyalkylene glycols such as poly(ethylene glycol) (PEG), polyalkylene oxides (PEO), polyalkylene terephthalates such as poly(ethylene terephthalate), polyvinyl alcohols (PVA), polyvinyl ethers, polyvinyl esters such as poly(vinyl acetate), polyvinyl halides such as poly(vinyl chloride) (PVC), polyvinylpyrrolidone, polysiloxanes, polystyrene (PS), polyurethanes, derivatized celluloses such as alkyl celluloses, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, hydroxypropylcellulose, carboxymethylcellulose, polymers of acrylic acids, such as poly(methyl(meth)acrylate) (PMMA), poly(ethyl(meth)acrylate), poly(butyl(meth)acrylate), poly(isobutyl(meth)acrylate), poly(hexyl(meth)acrylate), poly(isodecyl(meth)acrylate), poly(lauryl(meth)acrylate), poly(phenyl(meth)acrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate) and copolymers and mixtures thereof, polydioxanone and its copolymers, polyhydroxyalkanoates, polypropylene fumarate, polyoxymethylene, poloxamers, poly(ortho)esters, poly(butyric acid), poly(valeric acid), poly(lactide-co-caprolactone), and trimethylene carbonate, polyvinylpyrrolidone. The lipid nanoparticle may be coated or associated with a co-polymer such as, but not limited to, a block co-polymer, and (poly(ethylene glycol))-(poly(propylene oxide))-(poly(ethylene glycol)) triblock copolymer (see US Publication 20120121718 and US Publication 20100003337; herein incorporated by reference in their entireties). The co-polymer may be a polymer that is generally regarded as safe (GRAS) and the formation of the lipid nanoparticle may be in such a way that no new chemical entities are created. For example, the lipid nanoparticle may comprise poloxamers coating PLGA nanoparticles without forming new chemical entities which are still able to rapidly penetrate human mucus (Yang et al. Angew. Chem. Int. Ed. 2011 50:2597-2600; herein incorporated by reference in its entirety).

The vitamin of the polymer-vitamin conjugate may be vitamin E. The vitamin portion of the conjugate may be substituted with other suitable components such as, but not limited to, vitamin A, vitamin E, other vitamins, cholesterol, a hydrophobic moiety, or a hydrophobic component of other surfactants (e.g., sterol chains, fatty acids, hydrocarbon chains and alkylene oxide chains).

The lipid nanoparticle engineered to penetrate mucus may include surface altering agents such as, but not limited to, signal-sensor mmRNA, anionic protein (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as for example dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol and poloxamer), mucolytic agents (e.g., N-acetylcysteine, mugwort, bromelain, papain, clerodendrum, acetylcysteine, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin J34 dornase alfa, neltenexine, erdosteine) and various DNases including rhDNase. The surface altering agent may be embedded or enmeshed in the particle's surface or disposed (e.g., by coating, adsorption, covalent linkage, or other process) on the surface of the lipid nanoparticle. (see US Publication 20100215580 and US Publication 20080166414; herein incorporated by reference in their entireties).

The mucus penetrating lipid nanoparticles may comprise at least one signal-sensor mmRNA described herein. The signal-sensor mmRNA may be encapsulated in the lipid nanoparticle and/or disposed on the surface of the particle. The signal-sensor mmRNA may be covalently coupled to the lipid nanoparticle. Formulations of mucus penetrating lipid nanoparticles may comprise a plurality of nanoparticles. Further, the formulations may contain particles which may interact with the mucus and alter the structural and/or adhesive properties of the surrounding mucus to decrease mucoadhesion which may increase the delivery of the mucus penetrating lipid nanoparticles to the mucosal tissue.

Lipid nanoparticles may be engineered to alter the surface properties of particles so the lipid nanoparticles may penetrate the mucosal barrier. Mucus is located on mucosal tissue such as, but not limited to, oral (e.g., the buccal and esophageal membranes and tonsil tissue), ophthalmic, gastrointestinal (e.g., stomach, small intestine, large intestine, colon, rectum), nasal, respiratory (e.g., nasal, pharyngeal, tracheal and bronchial membranes), genital (e.g., vagin*l, cervical and urethral membranes). Nanoparticles larger than 10-200 nm which are preferred for higher drug encapsulation efficiency and the ability to provide the sustained delivery of a wide array of drugs have been thought to be too large to rapidly diffuse through mucosal barriers. Mucus is continuously secreted, shed, discarded or digested and recycled so most of the trapped particles may be removed from the mucosal tissue within seconds or within a few hours. Large polymeric nanoparticles (200 nm-500 nm in diameter) which have been coated densely with a low molecular weight polyethylene glycol (PEG) diffused through mucus only 4 to 6-fold lower than the same particles diffusing in water (Lai et al. PNAS 2007 104(5):1482-487; Lai et al. Adv Drug Deliv Rev. 2009 61(2): 158-171; herein incorporated by reference in their entirety). The transport of nanoparticles may be determined using rates of permeation and/or fluorescent microscopy techniques including, but not limited to, fluorescence recovery after photobleaching (FRAP) and high resolution multiple particle tracking (MPT).

The lipid nanoparticle engineered to penetrate mucus may comprise a polymeric material (i.e. a polymeric core) and/or a polymer-vitamin conjugate and/or a tri-block co-polymer. The polymeric material may including, but is not limited to, polyamines, polyethers, polyamides, polyesters, polycarbamates, polyureas, polycarbonates, poly(styrenes), polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyeneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates. The polymeric material may be biodegradable and/or biocompatible. Non-limiting examples of specific polymers include poly(caprolactone) (PCL), ethylene vinyl acetate polymer (EVA), poly(lactic acid) (PLA), poly(L-lactic acid) (PLLA), poly(glycolic acid) (PGA), poly(lactic acid-co-glycolic acid) (PLGA), poly(L-lactic acid-co-glycolic acid) (PLLGA), poly(D,L-lactide) (PDLA), poly(L-lactide) (PLLA), poly(D,L-lactide-co-caprolactone), poly(D,L-lactide-co-caprolactone-co-glycolide), poly(D,L-lactide-co-PEO-co-D,L-lactide), poly(D,L-lactide-co-PPO-co-D,L-lactide), polyalkyl cyanoacrylate, polyurethane, poly-L-lysine (PLL), hydroxypropyl methacrylate (HPMA), polyethyleneglycol, poly-L-glutamic acid, poly(hydroxy acids), polyanhydrides, polyorthoesters, poly(ester amides), polyamides, poly(ester ethers), polycarbonates, polyalkylenes such as polyethylene and polypropylene, polyalkylene glycols such as poly(ethylene glycol) (PEG), polyalkylene oxides (PEO), polyalkylene terephthalates such as poly(ethylene terephthalate), polyvinyl alcohols (PVA), polyvinyl ethers, polyvinyl esters such as poly(vinyl acetate), polyvinyl halides such as poly(vinyl chloride) (PVC), polyvinylpyrrolidone, polysiloxanes, polystyrene (PS), polyurethanes, derivatized celluloses such as alkyl celluloses, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, hydroxypropylcellulose, carboxymethylcellulose, polymers of acrylic acids, such as poly(methyl(meth)acrylate) (PMMA), poly(ethyl(meth)acrylate), poly(butyl(meth)acrylate), poly(isobutyl(meth)acrylate), poly(hexyl(meth)acrylate), poly(isodecyl(meth)acrylate), poly(lauryl(meth)acrylate), poly(phenyl(meth)acrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate) and copolymers and mixtures thereof, polydioxanone and its copolymers, polyhydroxyalkanoates, polypropylene fumarate, polyoxymethylene, poloxamers, poly(ortho)esters, poly(butyric acid), poly(valeric acid), poly(lactide-co-caprolactone), and trimethylene carbonate, polyvinylpyrrolidone. The lipid nanoparticle may be coated or associated with a co-polymer such as, but not limited to, a block co-polymer, and (poly(ethylene glycol))-(poly(propylene oxide))-(poly(ethylene glycol)) triblock copolymer (see US Publication 20120121718 and US Publication 20100003337; herein incorporated by reference in their entireties).

The vitamin of the polymer-vitamin conjugate may be vitamin E. The vitamin portion of the conjugate may be substituted with other suitable components such as, but not limited to, vitamin A, vitamin E, other vitamins, cholesterol, a hydrophobic moiety, or a hydrophobic component of other surfactants (e.g., sterol chains, fatty acids, hydrocarbon chains and alkylene oxide chains).

The lipid nanoparticle engineered to penetrate mucus may include surface altering agents such as, but not limited to, mmRNA, anionic protein (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as for example dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol and poloxamer), mucolytic agents (e.g., N-acetylcysteine, mugwort, bromelain, papain, clerodendrum, acetylcysteine, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin β4 dornase alfa, neltenexine, erdosteine) and various DNases including rhDNase. The surface altering agent may be embedded or enmeshed in the particle's surface or disposed (e.g., by coating, adsorption, covalent linkage, or other process) on the surface of the lipid nanoparticle. (see US Publication 20100215580 and US Publication 20080166414; herein incorporated by reference in their entireties).

The mucus penetrating lipid nanoparticles may comprise at least one signal-sensor polynucleotide, primary construct, or mmRNA described herein. The signal-sensor polynucleotide, primary construct, or mmRNA may be encapsulated in the lipid nanoparticle and/or disposed on the surface of the particle. The signal-sensor polynucleotide, primary construct, or mmRNA may be covalently coupled to the lipid nanoparticle. Formulations of mucus penetrating lipid nanoparticles may comprise a plurality of nanoparticles. Further, the formulations may contain particles which may interact with the mucus and alter the structural and/or adhesive properties of the surrounding mucus to decrease mucoadhesion which may increase the delivery of the mucus penetrating lipid nanoparticles to the mucosal tissue.

In one embodiment, the nanoparticle may be for a dual modality therapy such as described by Mieszawska et al. (Bioconjugate Chemistry, 2013, 24 (9), pp 1429-1434; the contents of which is herein incorporated by reference in its entirety) comprising at least one therapeutic agent (e.g., a signal-sequence polynucleotide described herein). The therapeutic agent or agents formulated in the lipid nanoparticle may be an anti-angiogenic and a cytotoxic agent (see e.g., the polymer-lipid nanoparticles taught by Mieszawska et al. Bioconjugate Chemistry, 2013, 24 (9), pp 1429-1434; the contents of which is herein incorporated by reference in its entirety).

In another embodiment, the nanoparticle may comprise a LyP-1 peptide such as the nanocarrier composition described in International Patent Publication No. WO2013100869, the contents of which are herein incorporated by reference in its entirety. The LyP-1 peptide may be contained in the nanoparticles disclosed herein, or may be a conjugate, derivative, analogue or pegylated form of the peptide. In one embodiment, a nanoparticle comprising the LyP-1 peptide may comprise a signal-sensor polynucleotide and may be used for cancer treatment and/or imaging.

In one embodiment, the signal-sensor polynucleotide, primary construct, or mmRNA is formulated as a lipoplex, such as, without limitation, the ATUPLEX™ system, the DACC system, the DBTC system and other siRNA-lipoplex technology from Silence Therapeutics (London, United Kingdom), STEMFECT™ from STEMGENT® (Cambridge, Mass.), and polyethylenimine (PEI) or protamine-based targeted and non-targeted delivery of nucleic acids (Aleku et al. Cancer Res. 2008 68:9788-9798; Strumberg et al. Int J Clin Pharmacol Ther 2012 50:76-78; Santel et al., Gene Ther 2006 13:1222-1234; Santel et al., Gene Ther 2006 13:1360-1370; Gutbier et al., Pulm Pharmacol. Ther. 2010 23:334-344; Kaufmann et al. Microvasc Res 2010 80:286-293 Weide et al. J Immunother. 2009 32:498-507; Weide et al. J Immunother. 2008 31:180-188; Pascolo Expert Opin. Biol. Ther. 4:1285-1294; Fotin-Mleczek et al., 2011 J. Immunother. 34:1-15; Song et al., Nature Biotechnol. 2005, 23:709-717; Peer et al., Proc Natl Acad Sci USA. 2007 6; 104:4095-4100; deFougerolles Hum Gene Ther. 2008 19:125-132; all of which are incorporated herein by reference in its entirety).

In one embodiment such formulations may also be constructed or compositions altered such that they passively or actively are directed to different cell types in vivo, including but not limited to hepatocytes, immune cells, tumor cells, endothelial cells, antigen presenting cells, and leukocytes (Akinc et al. Mol Ther. 2010 18:1357-1364; Song et al., Nat Biotechnol. 2005 23:709-717; Judge et al., J Clin Invest. 2009 119:661-673; Kaufmann et al., Microvasc Res 2010 80:286-293; Santel et al., Gene Ther 2006 13:1222-1234; Santel et al., Gene Ther 2006 13:1360-1370; Gutbier et al., Pulm Pharmacol. Ther. 2010 23:334-344; Basha et al., Mol. Ther. 2011 19:2186-2200; Fenske and Cullis, Expert Opin Drug Deliv. 2008 5:25-44; Peer et al., Science. 2008 319:627-630; Peer and Lieberman, Gene Ther. 2011 18:1127-1133; all of which are incorporated herein by reference in its entirety). One example of passive targeting of formulations to liver cells includes the DLin-DMA, DLin-KC2-DMA and MC3-based lipid nanoparticle formulations which have been shown to bind to apolipoprotein E and promote binding and uptake of these formulations into hepatocytes in vivo (Akinc et al. Mol Ther. 2010 18:1357-1364; herein incorporated by reference in its entirety). Formulations can also be selectively targeted through expression of different ligands on their surface as exemplified by, but not limited by, folate, transferrin, N-acetylgalactosamine (GalNAc), and antibody targeted approaches (Kolhatkar et al., Curr Drug Discov Technol. 2011 8:197-206; Musacchio and Torchilin, Front Biosci. 2011 16:1388-1412; Yu et al., Mol Membr Biol. 2010 27:286-298; Patil et al., Crit Rev Ther Drug Carrier Syst. 2008 25:1-61; Benoit et al., Biomacromolecules. 2011 12:2708-2714 Zhao et al., Expert Opin Drug Deliv. 2008 5:309-319; Akinc et al., Mol Ther. 2010 18:1357-1364; Srinivasan et al., Methods Mol Biol. 2012 820:105-116; Ben-Arie et al., Methods Mol Biol. 2012 757:497-507; Peer 2010 J Control Release. 20:63-68; Peer et al., Proc Natl Acad Sci USA. 2007 104:4095-4100; Kim et al., Methods Mol Biol. 2011 721:339-353; Subramanya et al., Mol Ther. 2010 18:2028-2037; Song et al., Nat Biotechnol. 2005 23:709-717; Peer et al., Science. 2008 319:627-630; Peer and Lieberman, Gene Ther. 2011 18:1127-1133; all of which are incorporated herein by reference in its entirety).

In one embodiment, the signal-sensor polynucleotide, primary construct, or mmRNA is formulated as a solid lipid nanoparticle. A solid lipid nanoparticle (SLN) may be spherical with an average diameter between 10 to 1000 nm. SLN possess a solid lipid core matrix that can solubilize lipophilic molecules and may be stabilized with surfactants and/or emulsifiers. In a further embodiment, the lipid nanoparticle may be a self-assembly lipid-polymer nanoparticle (see Zhang et al., ACS Nano, 2008, 2 (8), pp 1696-1702; herein incorporated by reference in its entirety).

Liposomes, lipoplexes, or lipid nanoparticles may be used to improve the efficacy of signal-sensor polynucleotide, primary construct, or mmRNA directed protein production as these formulations may be able to increase cell transfection by the signal-sensor polynucleotide, primary construct, or mmRNA; and/or increase the translation of encoded protein. One such example involves the use of lipid encapsulation to enable the effective systemic delivery of polyplex plasmid DNA (Heyes et al., Mol Ther. 2007 15:713-720; herein incorporated by reference in its entirety). The liposomes, lipoplexes, or lipid nanoparticles may also be used to increase the stability of the signal-sensor polynucleotide, primary construct, or mmRNA.

Polymers, Biodegradable Nanoparticles, and Core-Shell Nanoparticles

The signal-sensor polynucleotide, primary construct, and mmRNA of the invention can be formulated using natural and/or synthetic polymers. Non-limiting examples of polymers which may be used for delivery include, but are not limited to, Dynamic POLYCONJUGATE™ formulations from MIRUS® Bio (Madison, Wis.) and Roche Madison (Madison, Wis.), PHASERX™ polymer formulations such as, without limitation, SMARTT POLYMER TECHNOLOGY™ (Seattle, Wash.), DMRI/DOPE, poloxamer, VAXFECTIN® adjuvant from Vical (San Diego, Calif.), chitosan, cyclodextrin from Calando Pharmaceuticals (Pasadena, Calif.), dendrimers and poly(lactic-co-glycolic acid) (PLGA) polymers. RONDEL™ (RNAi/Oligonucleotide Nanoparticle Delivery) polymers (Arrowhead Research Corporation, Pasadena, Calif.) and pH responsive co-block polymers such as, but not limited to, PHASERX™ (Seattle, Wash.).

A non-limiting example of PLGA formulations include, but are not limited to, PLGA injectable depots (e.g., ELIGARD® which is formed by dissolving PLGA in 66% N-methyl-2-pyrrolidone (NMP) and the remainder being aqueous solvent and leuprolide. Once injected, the PLGA and leuprolide peptide precipitates into the subcutaneous space).

Many of these polymer approaches have demonstrated efficacy in delivering oligonucleotides in vivo into the cell cytoplasm (reviewed in deFougerolles Hum Gene Ther. 2008 19:125-132; herein incorporated by reference in its entirety). Two polymer approaches that have yielded robust in vivo delivery of nucleic acids, in this case with small interfering RNA (siRNA), are dynamic polyconjugates and cyclodextrin-based nanoparticles. The first of these delivery approaches uses dynamic polyconjugates and has been shown in vivo in mice to effectively deliver siRNA and silence endogenous target mRNA in hepatocytes (Rozema et al., Proc Natl Acad Sci USA. 2007 104:12982-12887). This particular approach is a multicomponent polymer system whose key features include a membrane-active polymer to which nucleic acid, in this case siRNA, is covalently coupled via a disulfide bond and where both PEG (for charge masking) and N-acetylgalactosamine (for hepatocyte targeting) groups are linked via pH-sensitive bonds (Rozema et al., Proc Natl Acad Sci USA. 2007 104:12982-12887). On binding to the hepatocyte and entry into the endosome, the polymer complex disassembles in the low-pH environment, with the polymer exposing its positive charge, leading to endosomal escape and cytoplasmic release of the siRNA from the polymer. Through replacement of the N-acetylgalactosamine group with a mannose group, it was shown one could alter targeting from asialoglycoprotein receptor-expressing hepatocytes to sinusoidal endothelium and Kupffer cells. Another polymer approach involves using transferrin-targeted cyclodextrin-containing polycation nanoparticles. These nanoparticles have demonstrated targeted silencing of the EWS-FLI1 gene product in transferrin receptor-expressing Ewing's sarcoma tumor cells (Hu-Lieskovan et al., Cancer Res. 2005 65: 8984-8982) and siRNA formulated in these nanoparticles was well tolerated in non-human primates (Heidel et al., Proc Natl Acad Sci USA 2007 104:5715-21). Both of these delivery strategies incorporate rational approaches using both targeted delivery and endosomal escape mechanisms.

The polymer formulation can permit the sustained or delayed release of signal-sensor polynucleotide, primary construct, or mmRNA (e.g., following intramuscular or subcutaneous injection). The altered release profile for the signal-sensor polynucleotide, primary construct, or mmRNA can result in, for example, translation of an encoded protein over an extended period of time. The polymer formulation may also be used to increase the stability of the signal-sensor polynucleotide, primary construct, or mmRNA. Biodegradable polymers have been previously used to protect nucleic acids other than mmRNA from degradation and been shown to result in sustained release of payloads in vivo (Rozema et al., Proc Natl Acad Sci USA. 2007 104:12982-12887; Sullivan et al., Expert Opin Drug Deliv. 2010 7:1433-1446; Convertine et al., Biomacromolecules. 2010 Oct. 1; Chu et al., Acc Chem Res. 2012 Jan. 13; Manganiello et al., Biomaterials. 2012 33:2301-2309; Benoit et al., Biomacromolecules. 2011 12:2708-2714; Singha et al., Nucleic Acid Ther. 2011 2:133-147; deFougerolles Hum Gene Ther. 2008 19:125-132; Schaffert and Wagner, Gene Ther. 2008 16:1131-1138; Chaturvedi et al., Expert Opin Drug Deliv. 2011 8:1455-1468; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464:1067-1070; herein incorporated by reference in its entirety).

In one embodiment, the pharmaceutical compositions may be sustained release formulations. In a further embodiment, the sustained release formulations may be for subcutaneous delivery. Sustained release formulations may include, but are not limited to, PLGA microspheres, ethylene vinyl acetate (EVAc), poloxamer, GELSITE® (Nanotherapeutics, Inc. Alachua, Fla.), HYLENEX® (Halozyme Therapeutics, San Diego Calif.), surgical sealants such as fibrinogen polymers (Ethicon Inc. Cornelia, Ga.). TISSELL® (Baxter International, Inc Deerfield, Ill.), PEG-based sealants, and COSEAL® (Baxter International, Inc Deerfield, Ill.).

As a non-limiting example modified mRNA may be formulated in PLGA microspheres by preparing the PLGA microspheres with tunable release rates (e.g., days and weeks) and encapsulating the signal-sensor modified mRNA in the PLGA microspheres while maintaining the integrity of the signal-sensor modified mRNA during the encapsulation process. EVAc are non-biodegradable, biocompatible polymers which are used extensively in pre-clinical sustained release implant applications (e.g., extended release products Ocusert a pilocarpine ophthalmic insert for glaucoma or progestasert a sustained release progesterone intrauterine device; transdermal delivery systems Testoderm, Duragesic and Selegiline; catheters). Poloxamer F-407 NF is a hydrophilic, non-ionic surfactant triblock copolymer of polyoxyethylene-polyoxypropylene-polyoxyethylene having a low viscosity at temperatures less than 5° C. and forms a solid gel at temperatures greater than 15° C. PEG-based surgical sealants comprise two synthetic PEG components mixed in a delivery device which can be prepared in one minute, seals in 3 minutes and is reabsorbed within 30 days. GELSITE® and natural polymers are capable of in-situ gelation at the site of administration. They have been shown to interact with protein and peptide therapeutic candidates through ionic interaction to provide a stabilizing effect.

Polymer formulations can also be selectively targeted through expression of different ligands as exemplified by, but not limited by, folate, transferrin, and N-acetylgalactosamine (GalNAc) (Benoit et al., Biomacromolecules. 2011 12:2708-2714; Rozema et al., Proc Natl Acad Sci USA. 2007 104:12982-12887; Davis, Mol Pharm. 2009 6:659-668; Davis, Nature 2010 464:1067-1070; herein incorporated by reference in its entirety).

The signal-sensor mmRNA of the invention may be formulated with or in a polymeric compound. The polymer may include at least one polymer such as, but not limited to, polyethylene glycol (PEG), poly(l-lysine)(PLL), PEG grafted to PLL, cationic lipopolymer, biodegradable cationic lipopolymer, polyethyleneimine (PEI), cross-linked branched poly(alkylene imines), a polyamine derivative, a modified poloxamer, a biodegradable polymer, biodegradable block copolymer, biodegradable random copolymer, biodegradable polyester copolymer, biodegradable polyester block copolymer, biodegradable polyester block random copolymer, linear biodegradable copolymer, poly[α-(4-aminobutyl)-L-glycolic acid) (PAGA), biodegradable cross-linked cationic multi-block copolymers or combinations thereof.

As a non-limiting example, the signal-sensor mmRNA of the invention may be formulated with the polymeric compound of PEG grafted with PLL as described in U.S. Pat. No. 6,177,274 herein incorporated by reference in its entirety. The formulation may be used for transfecting cells in vitro or for in vivo delivery of the signal-sensor mmRNA. In another example, the signal-sensor mmRNA may be suspended in a solution or medium with a cationic polymer, in a dry pharmaceutical composition or in a solution that is capable of being dried as described in U.S. Pub. Nos. 20090042829 and 20090042825 each of which are herein incorporated by reference in their entireties.

A polyamine derivative may be used to deliver nucleic acids or to treat and/or prevent a disease or to be included in an implantable or injectable device (U.S. Pub. No. 20100260817 herein incorporated by reference in its entirety). As a non-limiting example, a pharmaceutical composition may include the signal-sensor mmRNA and the polyamine derivative described in U.S. Pub. No. 20100260817 (the contents of which are incorporated herein by reference in its entirety.

For example, the signal-sensor mmRNA of the invention may be formulated in a pharmaceutical compound including a poly(alkylene imine), a biodegradable cationic lipopolymer, a biodegradable block copolymer, a biodegradable polymer, or a biodegradable random copolymer, a biodegradable polyester block copolymer, a biodegradable polyester polymer, a biodegradable polyester random copolymer, a linear biodegradable copolymer, PAGA, a biodegradable cross-linked cationic multi-block copolymer or combinations thereof. The biodegradable cationic lipopolymer may be made my methods known in the art and/or described in U.S. Pat. No. 6,696,038, U.S. App. Nos. 20030073619 and 20040142474 which is herein incorporated by reference in their entireties. The poly(alkylene imine) may be made using methods known in the art and/or as described in U.S. Pub. No. 20100004315, herein incorporated by reference in its entirety. The biodegradable polymer, biodegradable block copolymer, the biodegradable random copolymer, biodegradable polyester block copolymer, biodegradable polyester polymer, or biodegradable polyester random copolymer may be made using methods known in the art and/or as described in U.S. Pat. Nos. 6,517,869 and 6,267,987, the contents of which are each incorporated herein by reference in its entirety. The linear biodegradable copolymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,652,886. The PAGA polymer may be made using methods known in the art and/or as described in U.S. Pat. No. 6,217,912 herein incorporated by reference in its entirety. The PAGA polymer may be copolymerized to form a copolymer or block copolymer with polymers such as but not limited to, poly-L-lysine, polyarginine, polyornithine, histones, avidin, protamines, polylactides and poly(lactide-co-glycolides). The biodegradable cross-linked cationic multi-block copolymers may be made my methods known in the art and/or as described in U.S. Pat. No. 8,057,821 or U.S. Pub. No. 2012009145 herein incorporated by reference in their entireties. For example, the multi-block copolymers may be synthesized using linear polyethyleneimine (LPEI) blocks which have distinct patterns as compared to branched polyethyleneimines. Further, the composition or pharmaceutical composition may be made by the methods known in the art, described herein, or as described in U.S. Pub. No. 20100004315 or U.S. Pat. Nos. 6,267,987 and 6,217,912 herein incorporated by reference in their entireties.

As described in U.S. Pub. No. 20100004313, herein incorporated by reference in its entirety, a gene delivery composition may include a nucleotide sequence and a poloxamer. For example, the signal-sensor mmRNA of the present invention may be used in a gene delivery composition with the poloxamer described in U.S. Pub. No. 20100004313.

In one embodiment, the polymer formulation of the present invention may be stabilized by contacting the polymer formulation, which may include a cationic carrier, with a cationic lipopolymer which may be covalently linked to cholesterol and polyethylene glycol groups. The polymer formulation may be contacted with a cationic lipopolymer using the methods described in U.S. Pub. No. 20090042829 herein incorporated by reference in its entirety. The cationic carrier may include, but is not limited to, polyethylenimine, poly(trimethylenimine), poly(tetramethylenimine), polypropyleneimine, aminoglycoside-polyamine, dideoxy-diamino-b-cyclodextrin, spermine, spermidine, poly(2-dimethylamino)ethyl methacrylate, poly(lysine), poly(histidine), poly(arginine), cationized gelatin, dendrimers, chitosan, 1,2-Dioleoyl-3-Trimethylammonium-Propane (DOTAP), N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), 1-[2-(oleoyloxy)ethyl]-2-oleyl-3-(2-hydroxyethyl)imidazolinium chloride (DOTIM), 2,3-dioleyloxy-N-[2(sperminecarboxamido)ethyl]-N,N-dimethyl-1-propanaminium trifluoroacetate (DOSPA), 3B-[N—(N′,N′-Dimethylaminoethane)-carbamoyl]Cholesterol Hydrochloride (DC-Cholesterol HCl) diheptadecylamidoglycyl spermidine (DOGS), N,N-distearyl-N,N-dimethylammonium bromide (DDAB), N-(1,2-dimyristyloxyprop-3-yl)-N,N-dimethyl-N-hydroxyethyl ammonium bromide (DMRIE), N,N-dioleyl-N,N-dimethylammonium chloride DODAC) and combinations thereof.

The signal-sensor polynucleotide, primary construct, and mmRNA of the invention can also be formulated as a nanoparticle using a combination of polymers, lipids, and/or other biodegradable agents, such as, but not limited to, calcium phosphate. Components may be combined in a core-shell, hybrid, and/or layer-by-layer architecture, to allow for fine-tuning of the nanoparticle so to delivery of the signal-sensor polynucleotide, primary construct and mmRNA may be enhanced (Wang et al., Nat Mater. 2006 5:791-796; Fuller et al., Biomaterials. 2008 29:1526-1532; DeKoker et al., Adv Drug Deliv Rev. 2011 63:748-761; Endres et al., Biomaterials. 2011 32:7721-7731; Su et al., Mol Pharm. 2011 Jun. 6; 8(3):774-87; herein incorporated by reference in its entirety).

Biodegradable calcium phosphate nanoparticles in combination with lipids and/or polymers have been shown to deliver signal-sensor polynucleotides, primary constructs and mmRNA in vivo. In one embodiment, a lipid coated calcium phosphate nanoparticle, which may also contain a targeting ligand such as anisamide, may be used to deliver the signal-sensor polynucleotide, primary construct and mmRNA of the present invention. For example, to effectively deliver siRNA in a mouse metastatic lung model a lipid coated calcium phosphate nanoparticle was used (Li et al., J Contr Rel. 2010 142: 416-421; Li et al., J Contr Rel. 2012 158:108-114; Yang et al., Mol Ther. 2012 20:609-615). This delivery system combines both a targeted nanoparticle and a component to enhance the endosomal escape, calcium phosphate, in order to improve delivery of the siRNA.

In one embodiment, calcium phosphate with a PEG-polyanion block copolymer may be used to deliver signal-sensor polynucleotides, primary constructs and mmRNA (Kazikawa et al., J Contr Rel. 2004 97:345-356; Kazikawa et al., J Contr Rel. 2006 111:368-370).

In one embodiment, a PEG-charge-conversional polymer (Pitella et al., Biomaterials. 2011 32:3106-3114) may be used to form a nanoparticle to deliver the signal-sensor polynucleotides, primary constructs and mmRNA of the present invention. The PEG-charge-conversional polymer may improve upon the PEG-polyanion block copolymers by being cleaved into a polycation at acidic pH, thus enhancing endosomal escape.

The use of core-shell nanoparticles has additionally focused on a high-throughput approach to synthesize cationic cross-linked nanogel cores and various shells (Siegwart et al., Proc Natl Acad Sci USA. 2011 108:12996-13001). The complexation, delivery, and internalization of the polymeric nanoparticles can be precisely controlled by altering the chemical composition in both the core and shell components of the nanoparticle. For example, the core-shell nanoparticles may efficiently deliver siRNA to mouse hepatocytes after they covalently attach cholesterol to the nanoparticle.

In one embodiment, a hollow lipid core comprising a middle PLGA layer and an outer neutral lipid layer containing PEG may be used to delivery of the signal-sensor polynucleotide, primary construct and mmRNA of the present invention. As a non-limiting example, in mice bearing a luciferase-expressing tumor, it was determined that the lipid-polymer-lipid hybrid nanoparticle significantly suppressed luciferase expression, as compared to a conventional lipoplex (Shi et al, Angew Chem Int Ed. 2011 50:7027-7031).

Peptides and Proteins

The signal-sensor polynucleotide, primary construct, and mmRNA of the invention can be formulated with peptides and/or proteins in order to increase transfection of cells by the polynucleotide, primary construct, or mmRNA. In one embodiment, peptides such as, but not limited to, cell penetrating peptides and proteins and peptides that enable intracellular delivery may be used to deliver pharmaceutical formulations. A non-limiting example of a cell penetrating peptide which may be used with the pharmaceutical formulations of the present invention includes a cell-penetrating peptide sequence attached to polycations that facilitates delivery to the intracellular space, e.g., HIV-derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides (see, e.g., Caron et al., Mol. Ther. 3(3):310-8 (2001); Langel, Cell-Penetrating Peptides: Processes and Applications (CRC Press, Boca Raton Fla., 2002); El-Andaloussi et al., Curr. Pharm. Des. 11(28):3597-611 (2003); and Deshayes et al., Cell. Mol. Life Sci. 62(16):1839-49 (2005), all of which are incorporated herein by reference). The compositions can also be formulated to include a cell penetrating agent, e.g., liposomes, which enhance delivery of the compositions to the intracellular space. signal-sensor polynucleotides, primary constructs, and mmRNA of the invention may be complexed to peptides and/or proteins such as, but not limited to, peptides and/or proteins from Aileron Therapeutics (Cambridge, Mass.) and Permeon Biologics (Cambridge, Mass.) in order to enable intracellular delivery (Cronican et al., ACS Chem. Biol. 2010 5:747-752; McNaughton et al., Proc. Natl. Acad. Sci. USA 2009 106:6111-6116; Sawyer, Chem Biol Drug Des. 2009 73:3-6; Verdine and Hilinski, Methods Enzymol. 2012; 503:3-33; all of which are herein incorporated by reference in its entirety).

In one embodiment, the cell-penetrating polypeptide may comprise a first domain and a second domain. The first domain may comprise a supercharged polypeptide. The second domain may comprise a protein-binding partner. As used herein, “protein-binding partner” includes, but are not limited to, antibodies and functional fragments thereof, scaffold proteins, or peptides. The cell-penetrating polypeptide may further comprise an intracellular binding partner for the protein-binding partner. The cell-penetrating polypeptide may be capable of being secreted from a cell where the signal-sensor polynucleotide, primary construct, or mmRNA may be introduced.

Formulations of the including peptides or proteins may be used to increase cell transfection by the signal-sensor polynucleotide, primary construct, or mmRNA, alter the biodistribution of the signal-sensor polynucleotide, primary construct, or mmRNA (e.g., by targeting specific tissues or cell types), and/or increase the translation of encoded protein.

Cells

The signal-sensor polynucleotide, primary construct, and mmRNA of the invention can be transfected ex vivo into cells, which are subsequently transplanted into a subject. As non-limiting examples, the pharmaceutical compositions may include red blood cells to deliver modified RNA to liver and myeloid cells, virosomes to deliver modified RNA in virus-like particles (VLPs), and electroporated cells such as, but not limited to, from MAXCYTE® (Gaithersburg, Md.) and from ERYTECH® (Lyon, France) to deliver modified RNA. Examples of use of red blood cells, viral particles and electroporated cells to deliver payloads other than mmRNA have been documented (Godfrin et al., Expert Opin Biol Ther. 2012 12:127-133; Fang et al., Expert Opin Biol Ther. 2012 12:385-389; Hu et al., Proc Natl Acad Sci USA. 2011 108:10980-10985; Lund et al., Pharm Res. 2010 27:400-420; Huckriede et al., J Liposome Res. 2007; 17:39-47; Cusi, Hum Vaccin. 2006 2:1-7; de Jonge et al., Gene Ther. 2006 13:400-411; all of which are herein incorporated by reference in its entirety).

Cell-based formulations of the signal-sensor polynucleotide, primary construct, and mmRNA of the invention may be used to ensure cell transfection (e.g., in the cellular carrier), alter the biodistribution of the signal-sensor polynucleotide, primary construct, or mmRNA (e.g., by targeting the cell carrier to specific tissues or cell types), and/or increase the translation of encoded oncology-related protein.

A variety of methods are known in the art and suitable for introduction of nucleic acid into a cell, including viral and non-viral mediated techniques. Examples of typical non-viral mediated techniques include, but are not limited to, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magnetofection, liposome mediated transfer, microinjection, microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer (DEAE-dextran, polyethylenimine, polyethylene glycol (PEG) and the like) or cell fusion.

The technique of sonoporation, or cellular sonication, is the use of sound (e.g., ultrasonic frequencies) for modifying the permeability of the cell plasma membrane. Sonoporation methods are known to those in the art and are used to deliver nucleic acids in vivo (Yoon and Park, Expert Opin Drug Deliv. 2010 7:321-330; Postema and Gilja, Curr Pharm Biotechnol. 2007 8:355-361; Newman and Bettinger, Gene Ther. 2007 14:465-475; all herein incorporated by reference in their entirety). Sonoporation methods are known in the art and are also taught for example as it relates to bacteria in US Patent Publication 20100196983 and as it relates to other cell types in, for example, US Patent Publication 20100009424, each of which are incorporated herein by reference in their entirety.

Electroporation techniques are also well known in the art and are used to deliver nucleic acids in vivo and clinically (Andre et at, Curr Gene Ther. 2010 10:267-280; Chiarella et al., Curr Gene Ther. 2010 10:281-286; Hojman, Curr Gene Ther. 2010 10:128-138; all herein incorporated by reference in their entirety). In one embodiment, signal-sensor polynucleotides, primary constructs or mmRNA may be delivered by electroporation as described in Example 12.

Hyaluronidase

The intramuscular or subcutaneous localized injection of signal-sensor polynucleotide, primary construct, or mmRNA of the invention can include hyaluronidase, which catalyzes the hydrolysis of hyaluronan. By catalyzing the hydrolysis of hyaluronan, a constituent of the interstitial barrier, hyaluronidase lowers the viscosity of hyaluronan, thereby increasing tissue permeability (Frost, Expert Opin. Drug Deliv. (2007) 4:427-440; herein incorporated by reference in its entirety). It is useful to speed their dispersion and systemic distribution of encoded proteins produced by transfected cells. Alternatively, the hyaluronidase can be used to increase the number of cells exposed to a signal-sensor polynucleotide, primary construct, or mmRNA of the invention administered intramuscularly or subcutaneously.

Nanoparticle Mimics

The signal-sensor polynucleotide, primary construct or mmRNA of the invention may be encapsulated within and/or absorbed to a nanoparticle mimic. A nanoparticle mimic can mimic the delivery function organisms or particles such as, but not limited to, pathogens, viruses, bacteria, fungus, parasites, prions and cells. As a non-limiting example the signal-sensor polynucleotide, primary construct or mmRNA of the invention may be encapsulated in a non-viron particle which can mimic the delivery function of a virus (see International Pub. No. WO2012006376 herein incorporated by reference in its entirety).

Nanotubes

The signal-sensor polynucleotides, primary constructs or mmRNA of the invention can be attached or otherwise bound to at least one nanotube such as, but not limited to, rosette nanotubes, rosette nanotubes having twin bases with a linker, carbon nanotubes and/or single-walled carbon nanotubes, The signal-sensor polynucleotides, primary constructs or mmRNA may be bound to the nanotubes through forces such as, but not limited to, steric, ionic, covalent and/or other forces.

In one embodiment, the nanotube can release one or more signal-sensor polynucleotides, primary constructs or mmRNA into cells. The size and/or the surface structure of at least one nanotube may be altered so as to govern the interaction of the nanotubes within the body and/or to attach or bind to the signal-sensor polynucleotides, primary constructs or mmRNA disclosed herein. In one embodiment, the building block and/or the functional groups attached to the building block of the at least one nanotube may be altered to adjust the dimensions and/or properties of the nanotube. As a non-limiting example, the length of the nanotubes may be altered to hinder the nanotubes from passing through the holes in the walls of normal blood vessels but still small enough to pass through the larger holes in the blood vessels of tumor tissue.

In one embodiment, at least one nanotube may also be coated with delivery enhancing compounds including polymers, such as, but not limited to, polyethylene glycol. In another embodiment, at least one nanotube and/or the signal-sensor polynucleotides, primary constructs or mmRNA may be mixed with pharmaceutically acceptable excipients and/or delivery vehicles.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA are attached and/or otherwise bound to at least one rosette nanotube. The rosette nanotubes may be formed by a process known in the art and/or by the process described in International Publication No. WO2012094304, herein incorporated by reference in its entirety. At least one signal-sensor polynucleotide, primary construct and/or mmRNA may be attached and/or otherwise bound to at least one rosette nanotube by a process as described in International Publication No. WO2012094304, herein incorporated by reference in its entirety, where rosette nanotubes or modules forming rosette nanotubes are mixed in aqueous media with at least one signal-sensor polynucleotide, primary construct and/or mmRNA under conditions which may cause at least one signal-sensor polynucleotide, primary construct or mmRNA to attach or otherwise bind to the rosette nanotubes.

Conjugates

The signal-sensor polynucleotides, primary constructs, and mmRNA of the invention include conjugates, such as a polynucleotide, primary construct, or mmRNA covalently linked to a carrier or targeting group, or including two encoding regions that together produce a fusion protein (e.g., bearing a targeting group and therapeutic protein or peptide).

The conjugates of the invention include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), high-density lipoprotein (HDL), or globulin); an carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); or a lipid. The ligand may also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid, an oligonucleotide (e.g. an aptamer). Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolide) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacrylic acid), N-isopropylacrylamide polymers, or polyphosphazene. Example of polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.

Representative U.S. patents that teach the preparation of polynucleotide conjugates, particularly to RNA, include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941; 6,294,664; 6,320,017; 6,576,752; 6,783,931; 6,900,297; 7,037,646; each of which is herein incorporated by reference in their entirety.

In one embodiment, the conjugate of the present invention may function as a carrier for the signal-sensor mmRNA of the present invention. The conjugate may comprise a cationic polymer such as, but not limited to, polyamine, polylysine, polyalkyleneamine, and polyethylenimine which may be grafted to with poly(ethylene glycol). As a non-limiting example, the conjugate may be similar to the polymeric conjugate and the method of synthesizing the polymeric conjugate described in U.S. Pat. No. 6,586,524 herein incorporated by reference in its entirety.

The conjugates can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell. A targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, biotin, an RGD peptide, an RGD peptide mimetic or an aptamer.

Targeting groups can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell. Targeting groups may also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-glucosamine multivalent mannose, multivalent fucose, or aptamers. The ligand can be, for example, a lipopolysaccharide, or an activator of p38 MAP kinase.

The targeting group can be any ligand that is capable of targeting a specific receptor. Examples include, without limitation, folate, GalNAc, galactose, mannose, mannose-6P, aptamers, integrin receptor ligands, chemokine receptor ligands, transferrin, biotin, serotonin receptor ligands, PSMA, endothelin, GCPII, somatostatin, LDL, and HDL ligands. In particular embodiments, the targeting group is an aptamer. The aptamer can be unmodified or have any combination of modifications disclosed herein.

In one embodiment, pharmaceutical compositions of the present invention may include chemical modifications such as, but not limited to, modifications similar to locked nucleic acids.

Representative U.S. patents that teach the preparation of locked nucleic acid (LNA) such as those from Santaris, include, but are not limited to, the following: U.S. Pat. Nos. 6,268,490; 6,670,461; 6,794,499; 6,998,484; 7,053,207; 7,084,125; and 7,399,845, each of which is herein incorporated by reference in its entirety.

Representative U.S. patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found, for example, in Nielsen et al., Science, 1991, 254, 1497-1500.

Some embodiments featured in the invention include signal-sensor polynucleotides, primary constructs or mmRNA with phosphorothioate backbones and oligonucleosides with other modified backbones, and in particular —CH2—NH—CH2—, —CH2—N(CH3)—O—CH2— [known as a methylene (methylimino) or MMI backbone], —CH2—O—N(CH3)—CH2—, —CH2—N(CH3)—N(CH3)—CH2— and —N(CH3)—CH2—CH2— [wherein the native phosphodiester backbone is represented as —O—P(O)2—O—CH2—] of the above-referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above-referenced U.S. Pat. No. 5,602,240. In some embodiments, the polynucleotides featured herein have morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.

Modifications at the 2′ position may also aid in delivery. Preferably, modifications at the 2′ position are not located in a polypeptide-coding sequence, i.e., not in a translatable region. Modifications at the 2′ position may be located in a 5′UTR, a 3′UTR and/or a tailing region. Modifications at the 2′ position can include one of the following at the 2′ position: H (i.e., 2′-deoxy); F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Exemplary suitable modifications include O[(CH2)nO]mCH3, O(CH2nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON[CH2)nCH3)]2, where n and m are from 1 to about 10. In other embodiments, the signal-sensor polynucleotides, primary constructs or mmRNA include one of the following at the 2′ position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmaco*kinetic properties, or a group for improving the pharmacodynamic properties, and other substituents having similar properties. In some embodiments, the modification includes a 2′-methoxyethoxy (2′-O—CH2CH2OCH3, also known as 2′-O-(2-methoxyethyl) or T-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group. Another exemplary modification is 2′-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, as described in examples herein below, and 2′-dimethylaminoethoxyethoxy (also known in the art as 2′-O-dimethylaminoethoxyethyl or 2′-DMAEOE), i.e., 2′-O—CH2—O—CH2—N(CH2)2, also described in examples herein below. Other modifications include 2′-methoxy (2′-OCH3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2) and 2′-fluoro (2′-F). Similar modifications may also be made at other positions, particularly the 3′ position of the sugar on the 3′ terminal nucleotide or in 2′-5′ linked dsRNAs and the 5′ position of 5′ terminal nucleotide. signal-sensor polynucleotides of the invention may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920 and each of which is herein incorporated by reference.

In still other embodiments, the signal-sensor polynucleotide, primary construct, or mmRNA is covalently conjugated to a cell penetrating polypeptide. The cell-penetrating peptide may also include a signal peptide sequence. The conjugates of the invention can be designed to have increased stability; increased cell transfection; and/or altered the biodistribution (e.g., targeted to specific tissues or cell types).

Self-Assembled Nucleic Acid Nanoparticles

Self-assembled nanoparticles have a well-defined size which may be precisely controlled as the nucleic acid strands may be easily reprogrammable. For example, the optimal particle size for a cancer-targeting nanodelivery carrier is 20-100 nm as a diameter greater than 20 nm avoids renal clearance and enhances delivery to certain tumors through enhanced permeability and retention effect. Using self-assembled nucleic acid nanoparticles a single uniform population in size and shape having a precisely controlled spatial orientation and density of cancer-targeting ligands for enhanced delivery. As a non-limiting example, oligonucleotide nanoparticles were prepared using programmable self-assembly of short DNA fragments and therapeutic siRNAs. These nanoparticles are molecularly identical with controllable particle size and target ligand location and density. The DNA fragments and siRNAs self-assembled into a one-step reaction to generate DNA/siRNA tetrahedral nanoparticles for targeted in vivo delivery. (Lee et al., Nature Nanotechnology 2012 7:389-393).

Excipients

Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro (Lippincott, Williams & Wilkins, Baltimore, Md., 2006; incorporated herein by reference) discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention.

In some embodiments, a pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure. In some embodiments, an excipient is approved for use in humans and for veterinary use. In some embodiments, an excipient is approved by United States Food and Drug Administration. In some embodiments, an excipient is pharmaceutical grade. In some embodiments, an excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.

Pharmaceutically acceptable excipients used in the manufacture of pharmaceutical compositions include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical compositions.

Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.

Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.

Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and VEEGUM® [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan monolaurate [TWEEN®20], polyoxyethylene sorbitan [TWEEN®60], polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], sorbitan monostearate [Span®60], sorbitan tristearate [Span®65], glyceryl monooleate, sorbitan monooleate [SPAN®80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [MYRJ®45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and SOLUTOL®), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. CREMOPHOR®), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [BRIJ®30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, PLUORINC®F 68, POLOXAMER®188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof.

Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol,); natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (VEEGUM®), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid; polymethacrylates; waxes; water; alcohol; etc.; and combinations thereof.

Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives. Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate. Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and/or thimerosal. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid. Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol. Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid. Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, GLYDANT PLUS®, PHENONIP®, methylparaben, GERMALL®115, GERMABEN®II, NEOLONE®, KATHON®, and/or EUXYL®.

Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethyl alcohol, etc., and/or combinations thereof.

Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc., and combinations thereof.

Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, Eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, Litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.

Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator.

Delivery

The present disclosure encompasses the delivery of signal-sensor polynucleotides, primary constructs or mmRNA for any of therapeutic, pharmaceutical, diagnostic or imaging by any appropriate route taking into consideration likely advances in the sciences of drug delivery. Delivery may be naked or formulated.

Naked Delivery

The signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be delivered to a cell naked. As used herein in, “naked” refers to delivering signal-sensor polynucleotides, primary constructs or mmRNA free from agents which promote transfection. For example, the polynucleotides, primary constructs or mmRNA delivered to the cell may contain no modifications. The naked signal-sensor polynucleotides, primary constructs or mmRNA may be delivered to the cell using routes of administration known in the art and described herein.

Formulated Delivery

The signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be formulated, using the methods described herein. The formulations may contain signal-sensor polynucleotides, primary constructs or mmRNA which may be modified and/or unmodified. The formulations may further include, but are not limited to, cell penetration agents, a pharmaceutically acceptable carrier, a delivery agent, a bioerodible or biocompatible polymer, a solvent, and a sustained-release delivery depot. The formulated signal-sensor polynucleotides, primary constructs or mmRNA may be delivered to the cell using routes of administration known in the art and described herein.

The compositions may also be formulated for direct delivery to an organ or tissue in any of several ways in the art including, but not limited to, direct soaking or bathing, via a catheter, by gels, powder, ointments, creams, gels, lotions, and/or drops, by using substrates such as fabric or biodegradable materials coated or impregnated with the compositions, and the like.

Administration

The signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection, (into the base of the penis), intravagin*l administration, intrauterine, extra-amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), or in ear drops. In specific embodiments, compositions may be administered in a way which allows them cross the blood-brain barrier, vascular barrier, or other epithelial barrier. Non-limiting routes of administration for the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention are described below.

Parenteral and Injectable Administration

Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain embodiments for parenteral administration, compositions are mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.

Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.

Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In order to prolong the effect of an active ingredient, it is often desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.

Rectal and vagin*l Administration

Compositions for rectal or vagin*l administration are typically suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vagin*l cavity and release the active ingredient.

Oral Administration

Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders (e.g. starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders (e.g. carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants (e.g. glycerol), disintegrating agents (e.g. agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate), solution retarding agents (e.g. paraffin), absorption accelerators (e.g. quaternary ammonium compounds), wetting agents (e.g. cetyl alcohol and glycerol monostearate), absorbents (e.g. kaolin and bentonite clay), and lubricants (e.g. talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.

Topical or Transdermal Administration

As described herein, compositions containing the signal-sensor polynucleotides, primary constructs or mmRNA of the invention may be formulated for administration topically. The skin may be an ideal target site for delivery as it is readily accessible. Gene expression may be restricted not only to the skin, potentially avoiding nonspecific toxicity, but also to specific layers and cell types within the skin.

The site of cutaneous expression of the delivered compositions will depend on the route of nucleic acid delivery. Three routes are commonly considered to deliver signal-sensor polynucleotides, primary constructs or mmRNA to the skin: (i) topical application (e.g. for local/regional treatment and/or oncology-related applications); (ii) intradermal injection (e.g. for local/regional treatment and/or oncology-related applications); and (iii) systemic delivery (e.g. for treatment of dermatologic diseases that affect both cutaneous and extracutaneous regions). Signal-sensor polynucleotides, primary constructs or mmRNA can be delivered to the skin by several different approaches known in the art. Most topical delivery approaches have been shown to work for delivery of DNA, such as but not limited to, topical application of non-cationic liposome-DNA complex, cationic liposome-DNA complex, particle-mediated (gene gun), puncture-mediated gene transfections, and viral delivery approaches. After delivery of the nucleic acid, gene products have been detected in a number of different skin cell types, including, but not limited to, basal keratinocytes, sebaceous gland cells, dermal fibroblasts and dermal macrophages.

In one embodiment, the invention provides for a variety of dressings (e.g., wound dressings) or bandages (e.g., adhesive bandages) for conveniently and/or effectively carrying out methods of the present invention. Typically dressing or bandages may comprise sufficient amounts of pharmaceutical compositions and/or signal-sensor polynucleotides, primary constructs or mmRNA described herein to allow a user to perform multiple treatments of a subject(s).

In one embodiment, the invention provides for the signal-sensor polynucleotides, primary constructs or mmRNA compositions to be delivered in more than one injection.

In one embodiment, before topical and/or transdermal administration at least one area of tissue, such as skin, may be subjected to a device and/or solution which may increase permeability. In one embodiment, the tissue may be subjected to an abrasion device to increase the permeability of the skin (see U.S. Patent Publication No. 20080275468, herein incorporated by reference in its entirety). In another embodiment, the tissue may be subjected to an ultrasound enhancement device. An ultrasound enhancement device may include, but is not limited to, the devices described in U.S. Publication No. 20040236268 and U.S. Pat. Nos. 6,491,657 and 6,234,990; herein incorporated by reference in their entireties. Methods of enhancing the permeability of tissue are described in U.S. Publication Nos. 20040171980 and 20040236268 and U.S. Pat. No. 6,190,315; herein incorporated by reference in their entireties.

In one embodiment, a device may be used to increase permeability of tissue before delivering formulations of modified mRNA described herein. The permeability of skin may be measured by methods known in the art and/or described in U.S. Pat. No. 6,190,315, herein incorporated by reference in its entirety. As a non-limiting example, a modified mRNA formulation may be delivered by the drug delivery methods described in U.S. Pat. No. 6,190,315, herein incorporated by reference in its entirety.

In another non-limiting example tissue may be treated with a eutectic mixture of local anesthetics (EMLA) cream before, during and/or after the tissue may be subjected to a device which may increase permeability. Katz et al. (Anesth Analg (2004); 98:371-76; herein incorporated by reference in its entirety) showed that using the EMLA cream in combination with a low energy, an onset of superficial cutaneous analgesia was seen as fast as 5 minutes after a pretreatment with a low energy ultrasound.

In one embodiment, enhancers may be applied to the tissue before, during, and/or after the tissue has been treated to increase permeability. Enhancers include, but are not limited to, transport enhancers, physical enhancers, and cavitation enhancers. Non-limiting examples of enhancers are described in U.S. Pat. No. 6,190,315, herein incorporated by reference in its entirety.

In one embodiment, a device may be used to increase permeability of tissue before delivering formulations of modified mRNA described herein, which may further contain a substance that invokes an immune response. In another non-limiting example, a formulation containing a substance to invoke an immune response may be delivered by the methods described in U.S. Publication Nos. 20040171980 and 20040236268; herein incorporated by reference in their entireties.

Dosage forms for topical and/or transdermal administration of a composition may include ointments, pastes, creams, lotions, gels, foams, powders, solutions, sprays, inhalants and/or patches. Generally, an active ingredient is admixed under sterile conditions with a pharmaceutically acceptable excipient and/or any needed preservatives and/or buffers as may be required. Additionally, the present invention contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing the compound in the proper medium. Alternatively or additionally, rate may be controlled by either providing a rate controlling membrane and/or by dispersing the compound in a polymer matrix and/or gel.

Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions.

Topically-administrable formulations may, for example, comprise from about 0.1% to about 10% (w/w) active ingredient, although the concentration of active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical administration may further comprise one or more of the additional ingredients described herein.

Penetration Enhancers

In one embodiment, the signal-sensor polynucleotides, primary construct and mmRNA of present invention may use various penetration enhancers to deliver the signal-sensor polynucleotides, primary construct and mmRNA to at least one area associated with one or more hyperproliferative diseases, disorders or conditions. Most drugs are present in solution in both ionized and nonionized forms. However, usually only lipid soluble or lipophilic drugs readily cross cell membranes. It has been discovered that even non-lipophilic drugs may cross cell membranes if the membrane to be crossed is treated with a penetration enhancer. In addition to aiding the diffusion of non-lipophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs.

Penetration enhancers may be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of the above mentioned classes of penetration enhancers are described below in greater detail. Combinations of penetration enhancer may also be encompassed by the scope of the present invention, for example, fatty acids/salts in combination with bile acids/salts. Other non-limiting examples of combinations of penetration enhancers include the combination of sodium salt of lauric acid, capric acid and UDCA.

Surfactants

In connection with the present invention, surfactants (or “surface-active agents”) are chemical entities which, when dissolved in an aqueous solution, reduce the surface tension of the solution or the interfacial tension between the aqueous solution and another liquid, with the result that absorption of the signal-sensor polynucleotides, primary constructs and mmRNA through the mucosa is enhanced. In addition to bile salts and fatty acids, these penetration enhancers include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether) (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92); and perfluorochemical emulsions, such as FC-43 (Takahashi et al., J. Pharm. Pharmacol., 1988, 40, 252).

Fatty Acids

Various fatty acids and their derivatives which act as penetration enhancers include, but are not limited to, oleic acid, lauric acid, capric acid (n-decanoic acid), myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, dicaprate, monoolein (1-monooleoyl-rac-glycerol), dilaurin, caprylic acid, arachidonic acid, glycerol 1-monocaprate, 1-dodecylazacycloheptan-2-one, acylcarnitines, acylcholines, C1-C10 alkyl esters thereof (e.g., methyl, isopropyl and t-butyl), and mono- and diglycerides thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.) (Lee et al., Critical Reviews in Therapeutic Drug Carryier Systems, 1991, p. 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; El Hariri et al., J. Pharm. Pharmacol., 1992, 44, 651-654).

Bile Salts

The physiological role of bile includes the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (Brunton, Chapter 38 in: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al. Eds., McGraw-Hill, New York, 1996, pp. 934-935). Various natural bile salts, and their synthetic derivatives, act as penetration enhancers. Thus the term “bile salts” includes any of the naturally occurring components of bile as well as any of their synthetic derivatives. The bile salts of the invention include, but are not limited to, cholic acid (or its pharmaceutically acceptable sodium salt, sodium cholate), dehydrocholic acid (sodium dehydrocholate), deoxycholic acid (sodium deoxycholate), glucholic acid (sodium glucholate), glycholic acid (sodium glycocholate), glycodeoxycholic acid (sodium glycodeoxycholate), taurocholic acid (sodium taurocholate), taurodeoxycholic acid (sodium taurodeoxycholate), chenodeoxycholic acid (sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25-dihydro-fusidate (STDHF), sodium glycodihydrofusidate and polyoxyethylene-9-lauryl ether (POE) (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Swinyard, Chapter 39 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990, pages 782-783; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; Yamamoto et al., J. Pharm. Exp. Ther., 1992, 263, 25; Yamash*ta et al., J. Pharm. Sci., 1990, 79, 579-583).

Chelating Agents

Chelating agents, as used in connection with the present invention, can be defined as compounds that remove metallic ions from solution by forming complexes therewith, with the result that absorption of signal-sensor polynucleotides, primary construct and mmRNA through the mucosa is enhanced. With regards to their use as penetration enhancers in the present invention, chelating agents have the added advantage of also serving as DNase inhibitors, as most characterized DNA nucleases require a divalent metal ion for catalysis and are thus inhibited by chelating agents (Jarrett, J. Chromatogr., 1993, 618, 315-339). Chelating agents of the invention include but are not limited to disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and hom*ovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of beta-diketones (enamines)(Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; Buur et al., J. Control Rel., 1990, 14, 43-51).

Non-Chelating Non-Surfactants

As used herein, non-chelating non-surfactant penetration enhancing compounds can be defined as compounds that demonstrate insignificant activity as chelating agents or as surfactants but that nonetheless enhance absorption of signal-sensor polynucleotides, primary construct and mmRNA through the alimentary mucosa (Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33). This class of penetration enhancers include, but are not limited to, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92); and non-steroidal anti-inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone (Yamash*ta et al., J. Pharm. Pharmacol., 1987, 39, 621-626).

Agents that enhance uptake of signal-sensor polynucleotides, primary construct and mmRNA at the cellular level may also be added to the pharmaceutical and other compositions of the present invention. For example, cationic lipids, such as lipofectin (Junichi et al, U.S. Pat. No. 5,705,188), cationic glycerol derivatives, and polycationic molecules, such as polylysine (Lollo et al., PCT Application WO 97/30731), are also known to enhance the cellular uptake of signal-sensor polynucleotides, primary construct and mmRNA.

Other agents may be utilized to enhance the penetration of the administered signal-sensor polynucleotides, primary construct and mmRNA, including glycols such as ethylene glycol and propylene glycol, pyrrols such as 2-pyrrol, azones, and terpenes such as limonene and menthone.

Depot Administration

As described herein, in some embodiments, the composition is formulated in depots for extended release. Generally, a specific organ or tissue (a “target tissue”) is targeted for administration.

In some aspects of the invention, the signal-sensor polynucleotides, primary constructs or mmRNA are spatially retained within or proximal to a target tissue. Provided are method of providing a composition to a target tissue of a mammalian subject by contacting the target tissue (which contains one or more target cells) with the composition under conditions such that the composition, in particular the nucleic acid component(s) of the composition, is substantially retained in the target tissue, meaning that at least 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the composition is retained in the target tissue. Advantageously, retention is determined by measuring the amount of the nucleic acid present in the composition that enters one or more target cells. For example, at least 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the nucleic acids administered to the subject are present intracellularly at a period of time following administration. For example, intramuscular injection to a mammalian subject is performed using an aqueous composition containing a ribonucleic acid and a transfection reagent, and retention of the composition is determined by measuring the amount of the ribonucleic acid present in the muscle cells.

Aspects of the invention are directed to methods of providing a composition to a target tissue of a mammalian subject, by contacting the target tissue (containing one or more target cells) with the composition under conditions such that the composition is substantially retained in the target tissue. The composition contains an effective amount of a signal-sensor polynucleotides, primary constructs or mmRNA such that the polypeptide of interest is produced in at least one target cell. The compositions generally contain a cell penetration agent, although “naked” nucleic acid (such as nucleic acids without a cell penetration agent or other agent) is also contemplated, and a pharmaceutically acceptable carrier.

In some circ*mstances, the amount of an oncology-related protein produced by cells in a tissue is desirably increased. Preferably, this increase in oncology-related protein production is spatially restricted to cells within the target tissue. Thus, provided are methods of increasing production of an oncology-related protein of interest in a tissue of a mammalian subject. A composition is provided that contains signal-sensor polynucleotides, primary constructs or mmRNA characterized in that a unit quantity of composition has been determined to produce the polypeptide of interest in a substantial percentage of cells contained within a predetermined volume of the target tissue.

In some embodiments, the composition includes a plurality of different signal-sensor polynucleotides, primary constructs or mmRNA, where one or more than one of the signal-sensor polynucleotides, primary constructs or mmRNA encodes an oncology-related polypeptide of interest. Optionally, the composition also contains a cell penetration agent to assist in the intracellular delivery of the composition. A determination is made of the dose of the composition required to produce the oncology-related polypeptide of interest in a substantial percentage of cells contained within the predetermined volume of the target tissue (generally, without inducing significant production of the oncology-related polypeptide of interest in tissue adjacent to the predetermined volume, or distally to the target tissue). Subsequent to this determination, the determined dose is introduced directly into the tissue of the mammalian subject.

In one embodiment, the invention provides for the signal-sensor polynucleotides, primary constructs or mmRNA to be delivered in more than one injection or by split dose injections.

In one embodiment, the invention may be retained near target tissue using a small disposable drug reservoir or patch pump. Non-limiting examples of patch pumps include those manufactured and/or sold by BD® (Franklin Lakes, N.J.), Insulet Corporation (Bedford, Mass.), SteadyMed Therapeutics (San Francisco, Calif.), Medtronic (Minneapolis, Minn.), UniLife (York, Pa.), Valeritas (Bridgewater, N.J.), and SpringLeaf Therapeutics (Boston, Mass.).

Pulmonary Administration

A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity. Such a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm. Such compositions are suitably in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and/or using a self propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container. Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nm and at least 95% of the particles by number have a diameter less than 7 nm. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.

Low boiling propellants generally include liquid propellants having a boiling point of below 65° F. at atmospheric pressure. Generally the propellant may constitute 50% to 99.9% (w/w) of the composition, and active ingredient may constitute 0.1% to 20% (w/w) of the composition. A propellant may further comprise additional ingredients such as a liquid non-ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient).

Pharmaceutical compositions formulated for pulmonary delivery may provide an active ingredient in the form of droplets of a solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising active ingredient, and may conveniently be administered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as methylhydroxybenzoate. Droplets provided by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm.

Intranasal, Nasal and Buccal Administration

Formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition. Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 μm to 500 μm. Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.

Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, 0.1% to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.

Ophthalmic Administration

A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in the form of eye drops including, for example, a 0.1/1.0% (w/w) solution and/or suspension of the active ingredient in an aqueous or oily liquid excipient. Such drops may further comprise buffering agents, salts, and/or one or more other of any additional ingredients described herein. Other ophthalmically-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and/or in a liposomal preparation. Ear drops and/or eye drops are contemplated as being within the scope of this invention.

Payload Administration: Detectable Agents and Therapeutic Agents

The signal-sensor polynucleotides, primary constructs or mmRNA described herein can be used in a number of different scenarios in which delivery of a substance (the “payload”) to a biological target is desired, for example delivery of detectable substances for detection of the target, or delivery of a therapeutic agent. Detection methods can include, but are not limited to, both imaging in vitro and in vivo imaging methods, e.g., immunohistochemistry, bioluminescence imaging (BLI), Magnetic Resonance Imaging (MRI), positron emission tomography (PET), electron microscopy, X-ray computed tomography, Raman imaging, optical coherence tomography, absorption imaging, thermal imaging, fluorescence reflectance imaging, fluorescence microscopy, fluorescence molecular tomographic imaging, nuclear magnetic resonance imaging, X-ray imaging, ultrasound imaging, photoacoustic imaging, lab assays, or in any situation where tagging/staining/imaging is required.

The signal-sensor polynucleotides, primary constructs or mmRNA can be designed to include both a linker and a payload in any useful orientation. For example, a linker having two ends is used to attach one end to the payload and the other end to the nucleobase, such as at the C-7 or C-8 positions of the deaza-adenosine or deaza-guanosine or to the N-3 or C-5 positions of cytosine or uracil. The signal-sensor polynucleotide of the invention can include more than one payload (e.g., a label and a transcription inhibitor), as well as a cleavable linker. In one embodiment, the modified nucleotide is a modified 7-deaza-adenosine triphosphate, where one end of a cleavable linker is attached to the C7 position of 7-deaza-adenine, the other end of the linker is attached to an inhibitor (e.g., to the C5 position of the nucleobase on a cytidine), and a label (e.g., Cy5) is attached to the center of the linker (see, e.g., compound 1 of A*pCp C5 Parg Capless in FIG. 5 and columns 9 and 10 of U.S. Pat. No. 7,994,304, incorporated herein by reference). Upon incorporation of the modified 7-deaza-adenosine triphosphate to an encoding region, the resulting signal-sensor polynucleotide having a cleavable linker attached to a label and an inhibitor (e.g., a polymerase inhibitor). Upon cleavage of the linker (e.g., with reductive conditions to reduce a linker having a cleavable disulfide moiety), the label and inhibitor are released. Additional linkers and payloads (e.g., therapeutic agents, detectable labels, and cell penetrating payloads) are described herein.

For example, the signal-sensor polynucleotides, primary constructs or mmRNA described herein can be used in reprogramming induced pluripotent stem cells (iPS cells), which can directly track cells that are transfected compared to total cells in the cluster. In another example, a drug that may be attached to the signal-sensor polynucleotides, primary constructs or mmRNA via a linker and may be fluorescently labeled can be used to track the drug in vivo, e.g. intracellularly. Other examples include, but are not limited to, the use of signal-sensor polynucleotides, primary constructs or mmRNA in reversible drug delivery into cells.

The signal-sensor polynucleotides, primary constructs or mmRNA described herein can be used in intracellular targeting of a payload, e.g., detectable or therapeutic agent, to specific organelle. Exemplary intracellular targets can include, but are not limited to, the nuclear localization for advanced mRNA processing, or a nuclear localization sequence (NLS) linked to the mRNA containing an inhibitor.

In addition, the signal-sensor polynucleotides, primary constructs or mmRNA described herein can be used to deliver therapeutic agents to cells or tissues, e.g., in living animals. For example, the signal-sensor polynucleotides, primary constructs or mmRNA described herein can be used to deliver highly polar chemotherapeutics agents to kill cancer cells. The signal-sensor polynucleotides, primary constructs or mmRNA attached to the therapeutic agent through a linker can facilitate member permeation allowing the therapeutic agent to travel into a cell to reach an intracellular target.

In another example, the signal-sensor polynucleotides, primary constructs or mmRNA can be attached to the polynucleotides, primary constructs or mmRNA a viral inhibitory peptide (VIP) through a cleavable linker. The cleavable linker can release the VIP and dye into the cell. In another example, the signal-sensor polynucleotides, primary constructs or mmRNA can be attached through the linker to an ADP-ribosylate, which is responsible for the actions of some bacterial toxins, such as cholera toxin, diphtheria toxin, and pertussis toxin. These toxin proteins are ADP-ribosyltransferases that modify target proteins in human cells. For example, cholera toxin ADP-ribosylates G proteins modifies human cells by causing massive fluid secretion from the lining of the small intestine, which results in life-threatening diarrhea.

In some embodiments, the payload may be a therapeutic agent such as a cytotoxin, radioactive ion, chemotherapeutic, or other therapeutic agent. A cytotoxin or cytotoxic agent includes any agent that may be detrimental to cells. Examples include, but are not limited to, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthracinedione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, e.g., maytansinol (see U.S. Pat. No. 5,208,020 incorporated herein in its entirety), rachelmycin (CC-1065, see U.S. Pat. Nos. 5,475,092, 5,585,499, and 5,846,545, all of which are incorporated herein by reference), and analogs or hom*ologs thereof. Radioactive ions include, but are not limited to iodine (e.g., iodine 125 or iodine 131), strontium 89, phosphorous, palladium, cesium, iridium, phosphate, cobalt, yttrium 90, samarium 153, and praseodymium. Other therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thiotepa chlorambucil, rachelmycin (CC-1065), melphalan, carmustine (BSNU), lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine, vinblastine, taxol and maytansinoids).

In some embodiments, the payload may be a detectable agent, such as various organic small molecules, inorganic compounds, nanoparticles, enzymes or enzyme substrates, fluorescent materials, luminescent materials (e.g., luminol), bioluminescent materials (e.g., luciferase, luciferin, and aequorin), chemiluminescent materials, radioactive materials (e.g., 18F, 67Ga, 81mKr, 82Rb, 111In, 123I, 133Xe, 201Tl, 125I, 35S, 14C, 3H, or99m Tc (e.g., as pertechnetate (technetate(VII), TcO4)), and contrast agents (e.g., gold (e.g., gold nanoparticles), gadolinium (e.g., chelated Gd), iron oxides (e.g., superparamagnetic iron oxide (SPIO), monocrystalline iron oxide nanoparticles (MIONs), and ultrasmall superparamagnetic iron oxide (USPIO)), manganese chelates (e.g., Mn-DPDP), barium sulfate, iodinated contrast media (iohexol), microbubbles, or perfluorocarbons). Such optically-detectable labels include for example, without limitation, 4-acetamido-4′-isothiocyanatostilbene-2,2′disulfonic acid; acridine and derivatives (e.g., acridine and acridine isothiocyanate); 5-(2′-aminoethyl)aminonaphthalene-1-sulfonic acid (EDANS); 4-amino-N-[3-vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino-1-naphthyl)maleimide; anthranilamide; BODIPY; Brilliant Yellow; coumarin and derivatives (e.g., coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), and 7-amino-4-trifluoromethylcoumarin (Coumarin 151)); cyanine dyes; cyanosine; 4′,6-diaminidino-2-phenylindole (DAPI); 5′5″-dibromopyrogallol-sulfonaphthalein (Bromopyrogallol Red); 7-diethylamino-3-(4′-isothiocyanatophenyl)-4-methylcoumarin; diethylenetriamine pentaacetate; 4,4′-diisothiocyanatodihydro-stilbene-2,2′-disulfonic acid; 4,4′-diisothiocyanatostilbene-2,2′-disulfonic acid; 5-[dimethylamino]-naphthalene-1-sulfonyl chloride (DNS, dansylchloride); 4-dimethylaminophenylazophenyl-4′-isothiocyanate (DABITC); eosin and derivatives (e.g., eosin and eosin isothiocyanate); erythrosin and derivatives (e.g., erythrosin B and erythrosin isothiocyanate); ethidium; fluorescein and derivatives (e.g., 5-carboxyfluorescein (FAM), 5-(4,6-dichlorotriazin-2-yl)aminofluorescein (DTAF), 2′,7′-dimethoxy-4′5′-dichloro-6-carboxyfluorescein, fluorescein, fluorescein isothiocyanate, X-rhodamine-5-(and 6)-isothiocyanate (QFITC or XRITC), and fluorescamine); 2-[2-[3-[[1,3-dihydro-1,1-dimethyl-3-(3-sulfopropyl)-2H-benz[e]indol-2-ylidene]ethylidene]-2-[4-(ethoxycarbonyl)-1-piperazinyl]-1-cyclopenten-1-yl]ethenyl]-1,1-dimethyl-3-(3-sulfopropyl)-1H-benz[e]indolium hydroxide, inner salt, compound with n,n-diethylethanamine(1:1) (IR144); 5-chloro-2-[2-[3-[(5-chloro-3-ethyl-2(3H)-benzothiazol-ylidene)ethylidene]-2-(diphenylamino)-1-cyclopenten-1-yl]ethenyl]-3-ethyl benzothiazolium perchlorate (IR140); Malachite Green isothiocyanate; 4-methylumbelliferone orthocresolphthalein; nitrotyrosine; pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and derivatives (e.g., pyrene, pyrene butyrate, and succinimidyl 1-pyrene); butyrate quantum dots; Reactive Red 4 (CIBACRON™ Brilliant Red 3B-A); rhodamine and derivatives (e.g., 6-carboxy-X-rhodarnine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride rhodarnine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, sulforhodamine B, sulforhodamine 101, sulfonyl chloride derivative of sulforhodamine 101 (Texas Red), N,N,N′,N′tetramethyl-6-carboxyrhodamine (TAMRA) tetramethyl rhodamine, and tetramethyl rhodamine isothiocyanate (TRITC)); riboflavin; rosolic acid; terbium chelate derivatives; Cyanine-3 (Cy3); Cyanine-5 (Cy5); cyanine-5.5 (Cy5.5), Cyanine-7 (Cy7); IRD 700; IRD 800; Alexa 647; La Jolta Blue; phthalo cyanine; and naphthalo cyanine.

In some embodiments, the detectable agent may be a non-detectable pre-cursor that becomes detectable upon activation (e.g., fluorogenic tetrazine-fluorophore constructs (e.g., tetrazine-BODIPY FL, tetrazine-Oregon Green 488, or tetrazine-BODIPY TMR-X) or enzyme activatable fluorogenic agents (e.g., PROSENSE® (VisEn Medical))). In vitro assays in which the enzyme labeled compositions can be used include, but are not limited to, enzyme linked immunosorbent assays (ELISAs), immunoprecipitation assays, immunofluorescence, enzyme immunoassays (EIA), radioimmunoassays (RIA), and Western blot analysis. Combinations

The signal-sensor polynucleotides, primary constructs or mmRNA may be used in combination with one or more other therapeutic, prophylactic, diagnostic, or imaging agents. By “in combination with,” it is not intended to imply that the agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope of the present disclosure. Compositions can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. In some embodiments, the present disclosure encompasses the delivery of pharmaceutical, prophylactic, diagnostic, or imaging compositions in combination with agents that may improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body. As a non-limiting example, the signal-sensor nucleic acids or mmRNA may be used in combination with a pharmaceutical agent for the treatment of cancer or to control hyperproliferative cells. In U.S. Pat. No. 7,964,571, herein incorporated by reference in its entirety, a combination therapy for the treatment of solid primary or metastasized tumor is described using a pharmaceutical composition including a DNA plasmid encoding for interleukin-12 with a lipopolymer and also administering at least one anticancer agent or chemotherapeutic. Further, the signal-sensor nucleic acids and mmRNA of the present invention that encodes anti-proliferative molecules may be in a pharmaceutical composition with a lipopolymer (see e.g., U.S. Pub. No. 20110218231, herein incorporated by reference in its entirety, claiming a pharmaceutical composition comprising a DNA plasmid encoding an anti-proliferative molecule and a lipopolymer) which may be administered with at least one chemotherapeutic or anticancer agent.

Dosing

The present invention provides methods comprising administering modified mRNAs and their encoded proteins or complexes in accordance with the invention to a subject in need thereof. Nucleic acids, proteins or complexes, or pharmaceutical, imaging, diagnostic, or prophylactic compositions thereof, may be administered to a subject using any amount and any route of administration effective for preventing, treating, diagnosing, or imaging a disease, disorder, and/or condition (e.g., a disease, disorder, and/or condition relating to working memory deficits). The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular composition, its mode of administration, its mode of activity, and the like. Compositions in accordance with the invention are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention may be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective, prophylactically effective, or appropriate imaging dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.

In certain embodiments, compositions in accordance with the present invention may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.001 mg/kg to about 0.05 mg/kg, from about 0.005 mg/kg to about 0.05 mg/kg, from about 0.001 mg/kg to about 0.005 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging effect. The desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).

According to the present invention, it has been discovered that administration of mmRNA in split-dose regimens produce higher levels of proteins in mammalian subjects. As used herein, a “split dose” is the division of single unit dose or total daily dose into two or more doses, e.g, two or more administrations of the single unit dose. As used herein, a “single unit dose” is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single administration event. As used herein, a “total daily dose” is an amount given or prescribed in 24 hr period. It may be administered as a single unit dose. In one embodiment, the mmRNA of the present invention are administered to a subject in split doses. The mmRNA may be formulated in buffer only or in a formulation described herein.

Dosage Forms

A pharmaceutical composition described herein can be formulated into a dosage form described herein, such as a topical, intranasal, intratracheal, or injectable (e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous).

Liquid Dosage Forms

Liquid dosage forms for parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art including, but not limited to, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In certain embodiments for parenteral administration, compositions may be mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof.

Injectable

Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art and may include suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed include, but are not limited to, are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.

Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In order to prolong the effect of an active ingredient, it may be desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the signal-sensor polynucleotide, primary construct or mmRNA then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered signal-sensor polynucleotide, primary construct or mmRNA may be accomplished by dissolving or suspending the signal-sensor polynucleotide, primary construct or mmRNA in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the signal-sensor polynucleotide, primary construct or mmRNA in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of the signal-sensor polynucleotide, primary construct or mmRNA to polymer and the nature of the particular polymer employed, the rate of signal-sensor polynucleotide, primary construct or mmRNA release can be controlled. Examples of other biodegradable polymers include, but are not limited to, poly(orthoesters) and poly(anhydrides). Depot injectable formulations may be prepared by entrapping the signal-sensor polynucleotide, primary construct or mmRNA in liposomes or microemulsions which are compatible with body tissues.

Pulmonary

Formulations described herein as being useful for pulmonary delivery may also be use for intranasal delivery of a pharmaceutical composition. Another formulation suitable for intranasal administration may be a coarse powder comprising the active ingredient and having an average particle from about 0.2 μm to 500 μm. Such a formulation may be administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.

Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, contain about 0.1% to 20% (w/w) active ingredient, where the balance may comprise an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising active ingredient. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may have an average particle and/or droplet size in the range from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.

General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference).

Coatings or Shells

Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

Properties of Pharmaceutical Compositions

The pharmaceutical compositions described herein can be characterized by one or more of bioavailability, therapeutic window and/or volume of distribution.

Bioavailability

The signal-sensor polynucleotides, primary constructs or mmRNA, when formulated into a composition with a delivery agent as described herein, can exhibit an increase in bioavailability as compared to a composition lacking a delivery agent as described herein. As used herein, the term “bioavailability” refers to the systemic availability of a given amount of signal-sensor polynucleotides, primary constructs or mmRNA administered to a mammal. Bioavailability can be assessed by measuring the area under the curve (AUC) or the maximum serum or plasma concentration (Cmax) of the unchanged form of a compound following administration of the compound to a mammal. AUC is a determination of the area under the curve plotting the serum or plasma concentration of a compound along the ordinate (Y-axis) against time along the abscissa (X-axis). Generally, the AUC for a particular compound can be calculated using methods known to those of ordinary skill in the art and as described in G. S. Banker, Modern Pharmaceutics, Drugs and the Pharmaceutical Sciences, v. 72, Marcel Dekker, New York, Inc., 1996, herein incorporated by reference.

The Cmax value is the maximum concentration of the compound achieved in the serum or plasma of a mammal following administration of the compound to the mammal. The Cmax value of a particular compound can be measured using methods known to those of ordinary skill in the art. The phrases “increasing bioavailability” or “improving the pharmaco*kinetics,” as used herein mean that the systemic availability of a first signal-sensor polynucleotide, primary construct or mmRNA, measured as AUC, Cmax, or Cmin in a mammal is greater, when co-administered with a delivery agent as described herein, than when such co-administration does not take place. In some embodiments, the bioavailability of the signal-sensor polynucleotide, primary construct or mmRNA can increase by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%.

Therapeutic Window

The signal-sensor polynucleotides, primary constructs or mmRNA, when formulated into a composition with a delivery agent as described herein, can exhibit an increase in the therapeutic window of the administered signal-sensor polynucleotide, primary construct or mmRNA composition as compared to the therapeutic window of the administered signal-sensor polynucleotide, primary construct or mmRNA composition lacking a delivery agent as described herein. As used herein “therapeutic window” refers to the range of plasma concentrations, or the range of levels of therapeutically active substance at the site of action, with a high probability of eliciting a therapeutic effect. In some embodiments, the therapeutic window of the signal-sensor polynucleotide, primary construct or mmRNA when co-administered with a delivery agent as described herein can increase by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100%.

Volume of Distribution

The signal-sensor polynucleotides, primary constructs or mmRNA, when formulated into a composition with a delivery agent as described herein, can exhibit an improved volume of distribution (Vdist), e.g., reduced or targeted, relative to a composition lacking a delivery agent as described herein. The volume of distribution (Vdist) relates the amount of the drug in the body to the concentration of the drug in the blood or plasma. As used herein, the term “volume of distribution” refers to the fluid volume that would be required to contain the total amount of the drug in the body at the same concentration as in the blood or plasma: Vdist equals the amount of drug in the body/concentration of drug in blood or plasma. For example, for a 10 mg dose and a plasma concentration of 10 mg/L, the volume of distribution would be 1 liter. The volume of distribution reflects the extent to which the drug is present in the extravascular tissue. A large volume of distribution reflects the tendency of a compound to bind to the tissue components compared with plasma protein binding. In a clinical setting, Vdist can be used to determine a loading dose to achieve a steady state concentration. In some embodiments, the volume of distribution of the signal-sensor polynucleotide, primary construct or mmRNA when co-administered with a delivery agent as described herein can decrease at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%.

Biological Effect

In one embodiment, the biological effect of the signal-sensor modified mRNA delivered to the animals may be categorized by analyzing the protein expression in the animals. The protein expression may be determined from analyzing a biological sample collected from a mammal administered the signal-sensor modified mRNA of the present invention. In one embodiment, the expression protein encoded by the signal-sensor modified mRNA administered to the mammal of at least 50 pg/ml may be preferred. For example, a protein expression of 50-200 pg/ml for the protein encoded by the signal-sensor modified mRNA delivered to the mammal may be seen as a therapeutically effective amount of protein in the mammal.

Detection of Modified Nucleic Acids by Mass Spectrometry

Mass spectrometry (MS) is an analytical technique that can provide structural and molecular mass/concentration information on molecules after their conversion to ions. The molecules are first ionized to acquire positive or negative charges and then they travel through the mass analyzer to arrive at different areas of the detector according to their mass/charge (m/z) ratio.

Mass spectrometry is performed using a mass spectrometer which includes an ion source for ionizing the fractionated sample and creating charged molecules for further analysis. For example ionization of the sample may be performed by electrospray ionization (ESI), atmospheric pressure chemical ionization (APCI), photoionization, electron ionization, fast atom bombardment (FAB)/liquid secondary ionization (LSIMS), matrix assisted laser desorption/ionization (MALDI), field ionization, field desorption, thermospray/plasmaspray ionization, and particle beam ionization. The skilled artisan will understand that the choice of ionization method can be determined based on the analyte to be measured, type of sample, the type of detector, the choice of positive versus negative mode, etc.

After the sample has been ionized, the positively charged or negatively charged ions thereby created may be analyzed to determine a mass-to-charge ratio (i.e., m/z). Suitable analyzers for determining mass-to-charge ratios include quadrupole analyzers, ion traps analyzers, and time-of-flight analyzers. The ions may be detected using several detection modes. For example, selected ions may be detected (i.e., using a selective ion monitoring mode (SIM)), or alternatively, ions may be detected using a scanning mode, e.g., multiple reaction monitoring (MRM) or selected reaction monitoring (SRM).

Liquid chromatography-multiple reaction monitoring (LC-MS/MRM) coupled with stable isotope labeled dilution of peptide standards has been shown to be an effective method for protein verification (e.g., Keshishian et al., Mol Cell Proteomics 2009 8: 2339-2349; Kuhn et al., Clin Chem 2009 55:1108-1117; Lopez et al., Clin Chem 2010 56:281-290). Unlike untargeted mass spectrometry frequently used in biomarker discovery studies, targeted MS methods are peptide sequence-based modes of MS that focus the full analytical capacity of the instrument on tens to hundreds of selected peptides in a complex mixture. By restricting detection and fragmentation to only those peptides derived from proteins of interest, sensitivity and reproducibility are improved dramatically compared to discovery-mode MS methods. This method of mass spectrometry-based multiple reaction monitoring (MRM) quantitation of proteins can dramatically impact the discovery and quantitation of biomarkers via rapid, targeted, multiplexed protein expression profiling of clinical samples.

In one embodiment, a biological sample which may contain at least one protein encoded by at least one modified mRNA of the present invention may be analyzed by the method of MRM-MS. The quantification of the biological sample may further include, but is not limited to, isotopically labeled peptides or proteins as internal standards.

According to the present invention, the biological sample, once obtained from the subject, may be subjected to enzyme digestion. As used herein, the term “digest” means to break apart into shorter peptides. As used herein, the phrase “treating a sample to digest proteins” means manipulating a sample in such a way as to break down proteins in a sample. These enzymes include, but are not limited to, trypsin, endoproteinase Glu-C and chymotrypsin. In one embodiment, a biological sample which may contain at least one protein encoded by at least one modified mRNA of the present invention may be digested using enzymes.

In one embodiment, a biological sample which may contain protein encoded by modified mRNA of the present invention may be analyzed for protein using electrospray ionization. Electrospray ionization (ESI) mass spectrometry (ESIMS) uses electrical energy to aid in the transfer of ions from the solution to the gaseous phase before they are analyzed by mass spectrometry. Samples may be analyzed using methods known in the art (e.g., Ho et al., Clin Biochem Rev. 2003 24(1):3-12). The ionic species contained in solution may be transferred into the gas phase by dispersing a fine spray of charge droplets, evaporating the solvent and ejecting the ions from the charged droplets to generate a mist of highly charged droplets. The mist of highly charged droplets may be analyzed using at least 1, at least 2, at least 3 or at least 4 mass analyzers such as, but not limited to, a quadrupole mass analyzer. Further, the mass spectrometry method may include a purification step. As a non-limiting example, the first quadrupole may be set to select a single m/z ratio so it may filter out other molecular ions having a different m/z ratio which may eliminate complicated and time-consuming sample purification procedures prior to MS analysis.

In one embodiment, a biological sample which may contain protein encoded by modified mRNA of the present invention may be analyzed for protein in a tandem ESIMS system (e.g., MS/MS). As non-limiting examples, the droplets may be analyzed using a product scan (or daughter scan) a precursor scan (parent scan) a neutral loss or a multiple reaction monitoring.

In one embodiment, a biological sample which may contain protein encoded by modified mRNA of the present invention may be analyzed using matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MALDIMS). MALDI provides for the nondestructive vaporization and ionization of both large and small molecules, such as proteins. In MALDI analysis, the analyte is first co-crystallized with a large molar excess of a matrix compound, which may also include, but is not limited to, an ultraviolet absorbing weak organic acid. Non-limiting examples of matrices used in MALDI are α-cyano-4-hydroxycinnamic acid, 3,5-dimethoxy-4-hydroxycinnamic acid and 2,5-dihydroxybenzoic acid. Laser radiation of the analyte-matrix mixture may result in the vaporization of the matrix and the analyte. The laser induced desorption provides high ion yields of the intact analyte and allows for measurement of compounds with high accuracy. Samples may be analyzed using methods known in the art (e.g., Lewis, Wei and Siuzdak, Encyclopedia of Analytical Chemistry 2000:5880-5894). As non-limiting examples, mass analyzers used in the MALDI analysis may include a linear time-of-flight (TOF), a TOF reflectron or a Fourier transform mass analyzer.

In one embodiment, the analyte-matrix mixture may be formed using the dried-droplet method. A biologic sample is mixed with a matrix to create a saturated matrix solution where the matrix-to-sample ratio is approximately 5000:1. An aliquot (approximately 0.5-2.0 uL) of the saturated matrix solution is then allowed to dry to form the analyte-matrix mixture.

In one embodiment, the analyte-matrix mixture may be formed using the thin-layer method. A matrix hom*ogeneous film is first formed and then the sample is then applied and may be absorbed by the matrix to form the analyte-matrix mixture.

In one embodiment, the analyte-matrix mixture may be formed using the thick-layer method. A matrix hom*ogeneous film is formed with a nitro-cellulose matrix additive. Once the uniform nitro-cellulose matrix layer is obtained the sample is applied and absorbed into the matrix to form the analyte-matrix mixture.

In one embodiment, the analyte-matrix mixture may be formed using the sandwich method. A thin layer of matrix crystals is prepared as in the thin-layer method followed by the addition of droplets of aqueous trifluoroacetic acid, the sample and matrix. The sample is then absorbed into the matrix to form the analyte-matrix mixture.

The signal-sensor polynucleotides, primary constructs and mmRNA of the present invention are designed, in preferred embodiments, to provide for avoidance or evasion of deleterious bio-responses such as the immune response and/or degradation pathways, overcoming the threshold of expression and/or improving protein production capacity, improved expression rates or translation efficiency, improved drug or protein half life and/or protein concentrations, optimized protein localization, to improve one or more of the stability and/or clearance in tissues, receptor uptake and/or kinetics, cellular access by the compositions, engagement with translational machinery, secretion efficiency (when applicable), accessibility to circulation, and/or modulation of a cell's status, function and/or activity.

Therapeutics

Therapeutic Agents

The signal-sensor polynucleotides, primary constructs or mmRNA of the present invention, such as modified nucleic acids and modified RNAs, and the proteins translated from them described herein can be used as therapeutic or prophylactic agents. They are provided for use in medicine. For example, signal-sensor polynucleotide, primary construct or mmRNA described herein can be administered to a subject, wherein the signal-sensor polynucleotide, primary construct or mmRNA is translated in vivo to produce a therapeutic or prophylactic oncology-related polypeptide in the subject. Provided are compositions, methods, kits, and reagents for diagnosis, treatment or prevention of a disease or condition in humans and other mammals. The active therapeutic agents of the invention include signal-sensor polynucleotides, primary constructs or mmRNA, cells containing polynucleotides, primary constructs or mmRNA or polypeptides translated from the signal-sensor polynucleotides, primary constructs or mmRNA.

In certain embodiments, provided herein are combination therapeutics containing one or more signal-sensor polynucleotide, primary construct or mmRNA containing translatable regions that encode for a protein or proteins that boost a mammalian subject's immunity along with a protein that induces antibody-dependent cellular toxicity.

Provided herein are methods of inducing translation of a recombinant polypeptide in a cell population using the signal-sensor polynucleotide, primary construct or mmRNA described herein. Such translation can be in vivo, ex vivo, in culture, or in vitro. The cell population is contacted with an effective amount of a composition containing the signal-sensor nucleic acid that has at least one nucleoside modification, and a translatable region encoding the recombinant oncology-related polypeptide. The population is contacted under conditions such that the signal-sensor nucleic acid is localized into one or more cells of the cell population and the recombinant oncology-related polypeptide is translated in the cell from the signal-sensor nucleic acid.

An “effective amount” of the composition is provided based, at least in part, on the target tissue, target cell type, means of administration, physical characteristics of the nucleic acid (e.g., size, and extent of modified nucleosides), and other determinants. In general, an effective amount of the composition provides efficient protein production in the cell, preferably more efficient than a composition containing a corresponding unmodified nucleic acid. Increased efficiency may be demonstrated by increased cell transfection (i.e., the percentage of cells transfected with the nucleic acid), increased protein translation from the nucleic acid, decreased nucleic acid degradation (as demonstrated, e.g., by increased duration of protein translation from a modified nucleic acid), or reduced innate immune response of the host cell.

Aspects of the invention are directed to methods of inducing in vivo translation of a recombinant polypeptide in a mammalian subject in need thereof. Therein, an effective amount of a composition containing a nucleic acid that has at least one structural or chemical modification and a translatable region encoding the recombinant polypeptide is administered to the subject using the delivery methods described herein. The nucleic acid is provided in an amount and under other conditions such that the nucleic acid is localized into a cell of the subject and the recombinant polypeptide is translated in the cell from the nucleic acid. The cell in which the nucleic acid is localized, or the tissue in which the cell is present, may be targeted with one or more than one rounds of nucleic acid administration.

In certain embodiments, the administered signal-sensor polynucleotide, primary construct or mmRNA directs production of one or more recombinant polypeptides that provide a functional activity which is substantially absent in the cell, tissue or organism in which the recombinant oncology-related polypeptide is translated. For example, the missing functional activity may be enzymatic, structural, or gene regulatory in nature. In related embodiments, the administered signal-sensor polynucleotide, primary construct or mmRNA directs production of one or more recombinant oncology-related polypeptides that increases (e.g., synergistically) a functional activity which is present but substantially deficient in the cell in which the recombinant oncology-related polypeptide is translated.

In other embodiments, the administered signal-sensor polynucleotide, primary construct or mmRNA directs production of one or more recombinant polypeptides that replace an oncology-related polypeptide (or multiple oncology-related polypeptides) that is substantially absent in the cell in which the recombinant oncology-related polypeptide is translated. Such absence may be due to genetic mutation of the encoding gene or regulatory pathway thereof. In some embodiments, the recombinant oncology-related polypeptide increases the level of an endogenous oncology-related protein in the cell to a desirable level; such an increase may bring the level of the endogenous oncology-related protein from a subnormal level to a normal level or from a normal level to a super-normal level.

Alternatively, the recombinant oncology-related polypeptide functions to antagonize the activity of an endogenous protein present in, on the surface of, or secreted from the cell. Usually, the activity of the endogenous oncology-related protein is deleterious to the subject; for example, due to mutation of the endogenous oncology-related protein resulting in altered activity or localization. Additionally, the recombinant oncology-related polypeptide antagonizes, directly or indirectly, the activity of a biological moiety present in, on the surface of, or secreted from the cell. Examples of antagonized biological moieties include lipids (e.g., cholesterol), a lipoprotein (e.g., low density lipoprotein), a nucleic acid, a carbohydrate, a protein toxin such as shiga and tetanus toxins, or a small molecule toxin such as botulinum, cholera, and diphtheria toxins. Additionally, the antagonized biological molecule may be an endogenous protein that exhibits an undesirable activity, such as a cytotoxic or cytostatic activity.

The recombinant oncology-related proteins described herein may be engineered for localization within the cell, potentially within a specific compartment such as the nucleus, or are engineered for secretion from the cell or translocation to the plasma membrane of the cell.

In some embodiments, modified signal-sensor mRNAs and their encoded oncology-related polypeptides in accordance with the present invention may be used for treatment of any of a variety of diseases, disorders, and/or conditions described herein.

Oncology-Related Applications

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be used in the treatment, management, characterization and/or diagnosis of cancer, a cancer-related and/or a cancer treatment-related disorder, side effect and/or condition. Such disease, disorders and conditions include, but are not limited to, adrenal cortical cancer, advanced cancer, anal cancer, aplastic anemia, bileduct cancer, bladder cancer, bone cancer, bone metastasis, brain tumors, brain cancer, breast cancer, childhood cancer, cancer of unknown primary origin, Castleman disease, cervical cancer, colon/rectal cancer, endometrial cancer, esophagus cancer, Ewing family of tumors, eye cancer, fallopian tube cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gestational trophoblastic disease, Hodgkin disease, Kaposi sarcoma, renal cell carcinoma, laryngeal and hypopharyngeal cancer, acute lymphocytic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, chronic myelomonocytic leukemia, liver cancer, non-small cell lung cancer, small cell lung cancer, lung carcinoid tumor, lymphoma of the skin, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma in adult soft tissue, basal and squamous cell skin cancer, melanoma, small intestine cancer, stomach cancer, testicular cancer, thymus cancer, thyroid cancer, uterine sarcoma, vagin*l cancer, vulvar cancer, Waldenstrom macroglobulinemia, Wilms tumor.

In another embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be used in the treating, managing or manipulating at least one cancer-related or cancer treatment-related disorder, side effect or condition such as chemo brain, peripheral neuropathy, fatigue, depression, nausea and vomiting, pain, anemia, lymphedema, infections, second cancers caused by cancer treatment, sexual side effects, reduced fertility or infertility, ostomies, insomnia and hair loss.

In one embodiment, the signal-sensor polynucleotides, primary constructs and/or mmRNA may be used to reduce the effect of at least one symptom of cancer in a subject. The symptom may include, but is not limited to, weakness, aches and pains, fever, fatigue, weight loss, blood clots, increased blood calcium levels, low white blood cell count, short of breath, dizziness, headaches, hyperpigmentation, jaundice, erthema, pruritis, excessive hair growth, change in bowel habits, change in bladder function, long-lasting sores, white patches inside the mouth, white spots on the tongue, unusual bleeding or discharge, thickening or lump on parts of the body, indigestion, trouble swallowing, changes in warts or moles, change in new skin and nagging cough or hoarseness.

In one embodiment, the signal-sensor polynucleotides may be investigated in any number of cancer or normal cell lines. Non-limiting examples of cell lines which may be useful in these investigations include those from ATCC (Manassas, Va.) including MRC-5, A549, T84, NCI-H2126 [H2126], NCI-H1688 [H1688], WI-38, WI-38 VA-13 subline 2RA, WI-26 VA4, C3A [HepG2/C3A, derivative of Hep G2 (ATCC HB-8065)], THLE-3, H69AR, NCI-H292 [H292], CFPAC-1, NTERA-2 cl.D1 [NT2/D1], DMS 79, DMS 53, DMS 153, DMS 114, MSTO-211H, SW 1573 [SW-1573, SW1573], SW 1271 [SW-1271, SW1271], SHP-77, SNU-398, SNU-449, SNU-182, SNU-475, SNU-387, SNU-423, NL20, NL20-TA [NL20T-A], THLE-2, HBE135-E6E7, HCC827, HCC4006, NCI-H23 [H23], NCI-H1299, NCI-H187 [H187], NCI-H358 [H-358, H358], NCI-H378 [H378], NCI-H522 [H522], NCI-H526 [H526], NCI-H727 [H727], NCI-H810 [H810], NCI-H889 [H889], NCI-H1155 [H1155], NCI-H1404 [H1404], NCI-N87 [N87], NCI-H196 [H196], NCI-H211 [H211], NCI-H220 [H220], NCI-H250 [H250], NCI-H524 [H524], NCI-H647 [H647], NCI-H650 [H650], NCI-H711 [H711], NCI-H719 [H719], NCI-H740 [H740], NCI-H748 [H748], NCI-H774 [H774], NCI-H838 [H838], NCI-H841 [H841], NCI-H847 [H847], NCI-H865 [H865], NCI-H920 [H920], NCI-H1048 [H1048], NCI-H1092 [H1092], NCI-H1105 [H1105], NCI-H1184 [H1184], NCI-H1238 [H1238], NCI-H1341 [H1341], NCI-H1385 [H1385], NCI-H1417 [H1417], NCI-H1435 [H1435], NCI-H1436 [H1436], NCI-H1437 [H1437], NCI-H1522 [H1522], NCI-H1563 [H1563], NCI-H1568 [H1568], NCI-H1573 [H1573], NCI-H1581 [H1581], NCI-H1618 [H1618], NCI-H1623 [H1623], NCI-H1650 [H-1650, H1650], NCI-H1651 [H1651], NCI-H1666 [H-1666, H1666], NCI-H1672 [H1672], NCI-H1693 [H1693], NCI-H1694 [H1694], NCI-H1703 [H1703], NCI-H1734 [H-1734, 141734], NCI-H1755 [H1755], NCI-H1755 [H1755], NCI-H1770 [H1770], NCI-H1793 [H1793], NCI-H1836 [H1836], NCI-H1838 [H1838], NCI-H1869 [H1869], NCI-H1876 [H1876], NCI-H1882 [H1882], NCI-H1915 [H1915], NCI-H1930 [H1930], NCI-H1944 [H1944], NCI-H1975 [H-1975, H1975], NCI-H1993 [H1993], NCI-H2023 [H2023], NCI-H2029 [H2029], NCI-H2030 [H2030], NCI-H2066 [H2066], NCI-H2073 [H2073], NCI-H2081 [H2081], NCI-H2085 [H2085], NCI-H2087 [H2087], NCI-H2106 [H2106], NCI-H2110 [H2110], NCI-H2135 [H2135], NCI-H2141 [H2141], NCI-H2171 [H2171], NCI-H2172 [H2172], NCI-H2195 [H2195], NCI-H2196 [H2196], NCI-H2198 [H2198], NCI-H2227 [H2227], NCI-H2228 [H2228], NCI-H2286 [H2286], NCI-H2291 [H2291], NCI-H2330 [H2330], NCI-H2342 [H2342], NCI-H2347 [H2347], NCI-H2405 [H2405], NCI-H2444 [H2444], UMC-11, NCI-H64 [H64], NCI-H735 [H735], NCI-H735 [H735], NCI-H1963 [H1963], NCI-H2107 [H2107], NCI-H2108 [H2108], NCI-H2122 [H2122], Hs 573.T, Hs 573.Lu, PLC/PRF/5, BEAS-2B, Hep G2, Tera-1, Tera-2, NCI-H69 [H69], NCI-H128 [H128], ChaGo-K-1, NCI-H446 [H446], NCI-H209 [H209], NCI-H146 [H146], NCI-H441 [H441], NCI-H82 [H82], NCI-H460 [H460], NCI-H596 [H596], NCI-H676B [H676B], NCI-H345 [H345], NCI-H820 [H820], NCI-H520 [H520], NCI-H661 [H661], NCI-H510A [H510A, NCI-H510], SK-HEP-1, A-427, Calu-1, Calu-3, Calu-6, SK-LU-1, SK-MES-1, SW 900 [SW-900, SW900], Malme-3M, and Capan-1.

In one embodiment, the signal-sensor polynucleotides described herein may be investigated in human lung adenocarcinoma. As a non-limiting example, a signal-sensor polynucleotide encoding constitutively active caspase 3 fully modified with 5-methylcytidine and 1-methylpseudouridine or fully modified with 1-methylpseudouridine may be delivered to cultured human lung adenocarcinoma A549 cells (see e.g., the experiment outlined in Example 53). As another non-limiting example, a signal-sensor polynucleotide encoding constitutively active caspase 6 fully modified with 5-methylcytidine and 1-methylpseudouridine or fully modified with 1-methylpseudouridine may be delivered to cultured human lung adenocarcinoma A549 cells (see e.g., the experiment outlined in Example 53).

In another embodiment, the signal-sensor polynucleotides described herein may be investigated in human hepatocellular carcinoma. As a non-limiting example, a signal-sensor polynucleotide encoding constitutively active caspase 3 fully modified with 5-methylcytidine and 1-methylpseudouridine or fully modified with 1-methylpseudouridine may be delivered to human hepatocellular carcinoma Hep3B cells (see e.g., the experiment outlined in Example 54).

In one embodiment, the signal-sensor polynucleotides may be investigated in an animal model. As a non-limiting example, the animal model may be for lung cancer such as the lung cancer model of f*ckazawa et al (Anticancer Research, 2010; 30: 4193-4200) where a congenic mouse is created by crossing a ubiquitously expressing dominant negative Myc (Omomyc) mouse with a KRAS mutation-positive lung cancer model mouse. In the presence of Omomyc, lung tumors caused by the expression of mutated KRAS regresses in the congenic mouse, indicating that Omomyc caused tumor cell death of KRAS mutation-positive lung cancer.

As another non-limiting example, Human lung cancer xenografts are also prepared by the method of f*ckazawa where human lung cancer xenografts are established in 4-week-old female BALB/C nude mice (Charles River Laboratories Japan, Kanagawa, Japan) by subcutaneous inoculation of 4×106 A549 cells into the dorsal flank. The mice are randomly assigned into six groups (n=6/group). After the tumors reach a diameter of about 0.5 cm (approximately 6 days after tumor inoculations), each group of mice are injected with 100 μl solution containing PBS, 5×1010 vp of control or signal-sensor polynucleotide into the dorsal flank tumor for the selected dosing regimen. Animals are then observed closely and survival studies or other analyses are performed.

In one embodiment, the signal-sensor polynucleotides may be investigated in a transgenic animal model. As a non-limiting example, the transgenic animal model is a LSL-KRASG12D: TRE Omomyc:CMV rtTA triple transgenic model which involves the use of an adenovirus expressing Cre recombinase which is administered via inhalation to induce oncogene expression via excision of the foxed STOP codon, and ubiquitous Omomyc expression is controlled via doxycycline. The model is reported in Soucek et al. (Nature, 1-5 (2008)). As another non-limiting example, the mice of Soucek may be crossed with the LSLKRASG12D single transgenic mice (Jackson Laboratories) and may be used for inhalation delivered or otherwise lung-delivered studies of signal-sensor polynucleotides expressing MYC inhibitor D or other oncology related polypeptide described herein.

In another embodiment, the signal-sensor polynucleotides may be investigated in a mouse-in-mouse model such as, but not limited to a model which is akin to the p53−/−:c-Myc overexpressing HCC model of Zender (Cell. 2006 Jun. 30; 125(7): 1253-1267).

In one embodiment, the signal-sensor polynucleotides may be investigated in a Nongermline genetically engineered mouse model (NGEMM). As a non-limiting example, the design of mouse-in-mouse model may involve starting with the WT or tumor suppressor deleted (such as p53−/−) 129 Sv/Ev Mm ES cell clone; introduction of liver activated protein (LAP) promoter directed tetracycline transactivator (tTA) and tetO-luciferase for liver specific imaging; freezing the resulting LAP-tTA: tetO-luciferase clones to be used for c-Myc as well as other liver relevant programs oncogene; adding tetO driven oncogene, e.g. tetOcMyc; Freeze resulting LAP-tTA: tetO-luciferase: tetO-MYC clones; injecting resulting ES clones into C57BU6 blastocytes and implant in pseudo pregnant mothers whereby the resulting chimeric animals are the tumor model upon removal of doxycycline (i.e. Tet-Off). The type of model will ideally evince inducible nodules of c-Myc-driven, luciferase-expressing HCC surrounded by normal hepatocytes.

In another embodiment, the signal-sensor polynucleotides may be investigated in Orthotopic HCC models using the HEP3B cell lines in mice (Crown Bio).

As a non-limiting example, any of the animal models described above may be used to investigate a signal-sensor polynucleotide encoding MYC inhibitor D. The study may also include a signal-sensor polynucleotide encoding a negative control such as, but not limited to, an untranslatable mRNA for MYC inhibitor D and a vehicle only delivery. The animal may be evaluated for gene expression, tumor status and/or for any of the hallmarks that are generally associated with cancer phenotypes or genotypes.

As another non-limiting example, any of the animal models described above may be used to investigate a signal-sensor polynucleotide encoding dominant negative hTERT. The study may also include a signal-sensor polynucleotide encoding a negative control such as, but not limited to, an untranslatable mRNA for dominant negative hTERT and a vehicle only delivery. The animal may be evaluated for gene expression, tumor status and/or for any of the hallmarks that are generally associated with cancer phenotypes or genotypes.

As another non-limiting example, any of the animal models described above may be used to investigate a signal-sensor polynucleotide encoding dominant negative survivin. The study may also include a signal-sensor polynucleotide encoding a negative control such as, but not limited to, an untranslatable mRNA for dominant negative survivin and a vehicle only delivery. The animal may be evaluated for gene expression, tumor status and/or for any of the hallmarks that are generally associated with cancer phenotypes or genotypes.

In one embodiment, signal-sensor polynucleotides may include at least one miRNA-binding site in the 3′UTR in order to direct cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but not limited to, normal and/or cancerous cells in an animal model described herein. As a non-limiting example, a strong apoptotic signal and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR. As another non-limiting example, apoptosis inducing factor short isoform (AIFsh) and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR. As yet another non-limiting example, constitutively active (C.A.) caspase 6 and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR. As another non-limiting example, HSV1-tk and at least one miR-122a binding site is encoded by the signal-sensor polynucleotide where the at least one miR-122a binding site is located in the 3′UTR.

In another embodiment, signal-sensor polynucleotides may include three miRNA-binding sites in the 3′UTR in order to direct cytotoxic or cytoprotective mRNA therapeutics to specific cells such as, but not limited to, normal and/or cancerous cells in an animal model described herein. As a non-limiting example, a strong apoptotic signal and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR. As another non-limiting example, apoptosis inducing factor short isoform (AIFsh) and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR. As yet another non-limiting example, constitutively active (C.A.) caspase 6 and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR. As another non-limiting example, HSV1-tk and three miR-122a binding sites are encoded by the signal-sensor polynucleotide where the three miR-122a binding sites are located in the 3′UTR.

Common Categories of Cancer

Brain Cancer

Brain cancer is the growth of abnormal cells in the tissues of the brain usually related to the growth of malignant brain tumors. Brain tumors grow and press on the nearby areas of the brain which can stop that part of the brain from working the way it should. Brain cancer rarely spreads into other tissues outside of the brain. The grade of tumor, based on how abnormal the cancer cells look under a microscope, may be used to tell the difference between slow- and fast-growing tumors. Grade I tumors grow slowly, rarely spreads into nearby tissues, has cells that look like normal cells and the entire tumor may be removable by surgery. Grade II tumors also grow slowly but may spread into nearby tissue and may recur. Grade III tumors grow quickly, is likely to spread into nearby tissue and the tumor cells look very different from normal cells. Grade IV, high-grade, grows and spreads very quickly and there may be areas of dead cells in the tumor. Symptoms of brain cancer may include, but are not limited to, morning headache or headache that goes away after vomiting, frequent nausea and vomiting, vision, hearing, and speech problems, loss of balance and trouble walking, weakness on one side of the body, unusual sleepiness or change in activity level, unusual changes in personality or behavior, seizures.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with brain cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with brain cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with brain cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Breast Cancer

Breast cancer forms in the tissues of the breast, of both men and women, such as, but not limited to, the ducts and the lobules. The most common type of breast cancer is ductal carcinoma which begins in the cells of the ducts. Lobular cancer, which begins in the lobes or lobules, is often found in both breasts. An uncommon type of breast cancer, inflammatory breast cancer, causes the breast to be warm, red and swollen. Hereditary breast cancer makes up approximately 5-10% of all breast cancer and altered genes are common in some ethnic groups making that ethnic group more susceptible to breast cancer. Symptoms of breast cancer include, but are not limited to, a lump or thickening in or near the breast or in the underarm area, change in the size or shape of the breast, dimple or puckering in the skin of the breast, inward turned nipple of the breast, fluid from the nipple which is not breast milk, scaly, red or swollen skin on the breast, nipple, or areola, and dimples in the breast that look like the skin of orange (peau d'orange).

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with breast cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with breast cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with breast cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Cervical Cancer

Cervical cancer forms in the tissues of the cervix and is usually slow-growing. The cause of cervical cancer usually related to the human papillomavirus (NPV) infection. Although cervical cancer may not show any signs, possible symptoms may include, but are not limited to, vagin*l bleeding, unusual vagin*l discharge, pelvic pain and pain during sexual intercourse.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with cervical cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with cervical cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with cervical cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Esophageal Cancer

Esophageal cancer is cancer that forms in the tissues lining the esophagus. There are two common types of esophageal cancer which are named for the type of cells that become malignant. Squamous cell carcinoma is cancer that forms in the thin, flat cells lining the esophagus (also called epidermoid carcinoma). Cancer that begins in the glandular (secretory) cells which produce and release fluids such as mucus is called adenocarcinoma. Common symptoms associated with esophageal cancer include, but are not limited to, painful or difficult swallowing, weight loss, pain behind the breastbone, hoarseness and cough, and indigestion and heartburn.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with esophageal cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with esophageal cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with esophageal cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide.

Familial Cancer Syndrome

Familial cancer syndrome describes the genetic predisposition of a subject to develop cancer. 5-10% of all cancers are hereditary and are passed on through specific in specific genes passed from one blood relative to another. Subjects that inherit one of these gene changes may have a higher likelihood of developing cancer within their lifetime. Familial cancer syndrome includes disorder such as, but not limited to, Ataxia Telangiectasia, Basal Cell Nevus Syndrome, Nevoid Basal Cell Carcinoma Syndrome, Gorlin Syndrome, Beck-with Wiedemann Syndrome, Birt-Hogg-Dube Syndrome, Bloom Syndrome, hereditary breast and/or ovarian cancer, Carney Complex, Types I and II, Familial Chordoma, Colon Cancer, Hereditary Nonpolyposis-Lynch Syndrome, Costello Syndrome, Facio-Cutaneous-Skeletal Syndrome, Cowden Syndrome, Dyskeratosis Congenita, Tylosis with Esophageal Cancer, Keratosis Palmaris et Plantaris with Esophageal Cancer, Howel-Evans Syndrome, Hereditary Multiple Exostosis, Fanconi Anemia, Hereditary Diffuse Gastric Cancer, Gastrointestinal Stromal Tumor, Multiple Gastrointestinal Stromal Tumor, Familial Hyperparathyroidism, Acute Myeloid Leukemia, Familial Leukemia, Chronic Lymphocytic Leukemia, Li-Fraumeni Syndrome, Hodgkin Lymphoma, Non-Hodgkin Lymphoma, Hereditary Multiple Melanoma, Mosaic Varigated Aneuploidy, Multiple Endocrine Neoplasia Type I, Type 2A and 2B, Familial Medullary Thyroid Cancer, Familial Multiple Myeloma, Hereditary Neuroblastoma, Neurofibromatosis Type 1 and 2, Nijmegen Breakage Syndrome, Hereditary Pancreatic Cancer, Hereditary Paraganglioma, Peutz-Jeghers Syndrome, Familial Adenomatous Polyposis, Familial Juvenile Polyposis, MYH-Associated Polyposis, Hereditary Prostate Cancer, Hereditary Renal Cell Carcinoma with Multiple Cutaneous and Uterine Leiomyomas, Hereditary Renal Cell Carcinoma, Hereditary Papillary Renal Cell Carcinoma, Rhabdoid Predisposition Syndrome, Rothmund-Thomson Syndrome, Simpson-Golabi-Behmel Syndrome, Familial Testicular Germ Cell Tumor, Familial Non-medullary Thyroid Carcinoma, Tuberous Sclerosis Complex, von Hippel-Lindau Syndrome, Familial Waldenstrom Macroglobulinemia, Werner Syndrome, Familial Wilms Tumor and Xeroderma Pigmentosum.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with Familial cancer syndrome by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with Familial cancer syndrome by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with Familial cancer syndrome by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Leukemia

Leukemia is a form of cancer that starts in blood-forming tissue such as the bone marrow which can cause a large number of blood cells to be produced and enter the blood stream. Leukemia can also spread to the central nervous system and cause brain and spinal cord cancer. Types of leukemia include, but are not limited to, adult acute lymphoblastic, childhood acute lymphoblastic, adduct acute myeloid, chronic lymphocytic, chronic myelogenous and hairy cell. Non-limiting examples of symptoms of leukemia include weakness or feeling tired, fever, easy bruising or bleeding, petechiae, shortness of breath, weight loss or loss of appetite, pain in the bones or stomach, pain or feeling of fullness below the ribs, and painless lumps in the neck, underarm, stomach or groin.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with leukemia by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with leukemia by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with leukemia by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Liver Cancer

There are two types of liver cancer, primary liver cancer which forms in the tissue of the liver and secondary liver cancer, or metastatic liver cancer, that spreads to the liver from another part of the body. Possible symptoms of liver cancer include, but are not limited to, a hard lump on the right side just below the rib cage, discomfort in the upper abdomen on the right side, pain around the right shoulder blade, unexplained weight loss, jaundice, unusual tiredness, nausea and loss of appetite.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with liver cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with liver cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with liver cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Hepatocellular Carcinoma

The c-myc protein is a multifunctional bHLHZip transcription factor with critical roles in normal cellular processes and aberrantly regulated in the majority of human cancers. c-, N- and L-Myc are family members that can dimerize with partners such as Max, Mad and Miz-1. The protein is implicated in the transactivation and repression of a vast number of proposed transcriptional targets and recent work has demonstrated a role for Myc as “transcriptional amplifier” of otherwise transactivated genes in developing cancers. It has a well established function in cancer cell proliferation, growth, biosynthetic metabolism, ribogenesis and translation and possibly a non-redundant node through which oncogenic signals must navigate.

MYC inhibitor D (also known as Omomyc) is a unique dominant-negative 90 a.a. protein comprised of the human c-Myc oligomerization domain with 4 introduced mutations E57T, E641, R70Q, R71N (Soucek et al., Oncogene, 1998; 17, 2463-2472). Importantly, it exhibits selectivity in binding and inhibitory capability: binding c-Myc, N-Myc, Max and Miz-1. It also prevents E-box mediated transactivation while retaining Miz-1 directed transrepression. The therapeutic potential of MYC inhibitor D has been specifically exhibited in vivo where transgenic expression of OMOMYC blocked MycERTAM induced keratinocyte proliferation (Soucek et al., CDD 2004; 11, 1038-1045); transgenic Omomyc prevented the establishment and induced the regression of forming and mature lung tumors, respectively, in the LSL-KrasG12D mouse model with reversible toxicity (Soucek et al., Nature 2008, 455, 679-683); transgenic Omomyc prevents tumor formation and regresses established tumors in the RIP1-TAG2 model of pancreatic neuroendocrine cancer with controllable side effects, and further shows a role for cancer cell Myc in the maintenance of a permissive tumor microenvironment (Sodir et al., Genes and Development 2011, 25, 907-916); and it was reported “that Omomyc induces cell death of KRAS-mutated human lung adenocarcinoma A549 cells in vitro and in vivo” (f*ckazawa et al., Anticancer Res, 2010, 30, 4193-4200).

Although it stands to reason that the inhibition of oncogenic c-Myc via the directed expression of MYC inhibitor D might prove to be an effective therapy in at least a subset of HCCs, proof of concept in HCC remains to be demonstrated.

In some embodiments, the present invention includes signal-sensor polynucleotides encoding MYC inhibitor D as the oncology-related polypeptide; with or without a sensor sequence for the treatment of hepatocellular carcinoma (HCC). The studies of HCC may be performed in any of the subclasses of HCC cell lines as described by Hoshida et al (Cancer Research 2009; 69: 7385-7392). These include S2 cells which have higher TGF-beta and WNT signaling and demonstrate and associated with a greater risk of early recurrence, S2 which exhibit increased myc and AKT expression and the highest level of alpha feto-protein or S3 which retain the hepatocyte like phenotype. S1 and S2 types have also been shown to exhibit increased E2F1 and decreased p53 expression; while S2 alone has shown decreased levels of interferon. S1 cell lines include SNU-387, SNU-423, SNU-449, SNU-475, SNU-182, SK-Hep1, HLE, HLF, and Focus, whereas S2 cell lines include Huh-1, Huh-6, Huh-7, HepG2, Hep3B, Hep3B-TR, Hep40, and PLC/PRF/5 cells.

Lung Cancer

Lung cancer forms in the tissues of the lung usually in the cells lining the air passages and is classified as either small cell lung cancer or non-small cell lung cancer. There are two types of small cell lung cancer, small cell carcinoma and combined small cell carcinoma. The types of on-small cell lung cancer are squamous cell carcinoma (cancer begins in the squamous cells), large cell carcinoma (cancer may begin in several types of cells) and adenocarcinoma (cancer begins in the cells that line the alveoli and in cells that make mucus). Symptoms of lung cancer include, but are not limited to, chest discomfort or pain, cough that does not go away or gets worse over time, trouble breathing, wheezing, blood in the sputum, hoarseness, loss of appetite, weight loss for no known reason, feeling very tired, trouble swallowing and swelling in the face and/or veins in the neck.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with lung cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with lung cancer by administering to said subject an isolated polynucleotide encoding a polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with lung cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Lymphoma

Lymphoma is cancer that beings in the cells of the immune system. Subjects who have Hodgkin lymphoma have a cell called Reed-Sternberg cell and non-Hodgkin lymphoma includes a large group of cancers of immune system cells. Examples of Lymphoma include, but are not limited to, painless, swollen lymph nodes in the neck, underarm or groin, fever for no known reason, drenching night sweats, weight loss for no known reason, itchy skin and fatigue.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with lymphoma by administering to said subject an isolated polynucleotide encoding a polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with lymphoma by administering to said subject an isolated polynucleotide encoding a polypeptide of interest. In one embodiment, the polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with lymphoma by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Ovarian Cancer

Ovarian cancer is cancer which forms in the tissues of the ovary which are either ovarian epithelial carcinomas (begins on the surface of the ovary) or malignant germ cell tumors (cancer that begins in the egg cells). Symptoms of ovarian cancer include, but are not limited to, pain or swelling in the abdomen, pain in the pelvis, gastrointestinal problems such as gas, bloating, or constipation and vagin*l bleeding after menopause.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with ovarian cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or signal-sensor mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with ovarian cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or signal-sensor mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with ovarian cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Prostate Cancer

Prostate that forms in the tissue of the prostate mainly affects older men. Non-limiting examples of prostate cancer include, but are not limited to, weak or interrupted flow of urine, frequent urination, trouble urinating, pain or burning during urination, blood in the urine or sem*n, pain in the back, hips or pelvis that does not go away and painful ejacul*tion.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with prostate cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with prostate cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with prostate cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Testicular Cancer

Testicular cancer forms in the tissues of one or both testicl*s and is most common in young or middle-aged men. Most testicular cancers being in germ cells and are called testicular germ cell tumors. There are two types of testicular germ cell tumors called seminomas and nonseminomas. Common symptoms of testicular cancer include, but are not limited to, a painless lump or swelling in either testicl*, change in how the testicl* feels, dull ache in the lower abdomen or the groin, sudden build-up of fluid in the scrotum and pain or discomfort in a testicl* or in the scrotum.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with testicular cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with testicular cancer by administering to said subject an isolated signal-sensor polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with testicular cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Throat Cancer

Throat cancer forms in the tissues of the pharynx and includes cancer of the nasopharynx (nasopharyngeal cancer), oropharynx (oropharyngeal cancer), hypopharynx (hypopharyngeal cancer), and larynx (laryngeal cancer). Common symptoms of throat cancer include, but are not limited to, a sore throat that does not go away, ear pain, lump in the neck, painful or difficulty swallowing, change or hoarseness in the voice, trouble breathing or speaking, nosebleeds, trouble hearing, pain or ringing in the ear, headaches, dull pain behind the breast bone, cough and weight loss for no reason.

In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to treat a disease, disorder and/or condition in a subject who has been diagnosed or may be diagnosed with throat cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce, eliminate, or prevent tumor growth in a subject who has been diagnosed or may be diagnosed with throat cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest. In one embodiment, the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention may be used to reduce and/or ameliorate at least one symptom of cancer in a subject who has been diagnosed or may be diagnosed with throat cancer by administering to said subject an isolated polynucleotide encoding an oncology-related polypeptide of interest.

Inhibition of Hypoxia-Inducible Factors (HIFs)

Hypoxia-inducible factors (HIFs) control cellular adaptation to oxygen deprivation. Cancer cells engage HIFs to sustain their growth in adverse conditions, thus promoting a cellular reprograming that includes metabolism, proliferation, survival and mobility. HIFs overexpression in human cancer biopsies correlates with high metastasis and mortality.

HIFs regulate genes related to metabolism such as GLUT1, GLUT3, ALDOA, ENOI, GAPDH, HK1, HK2, PFKL, PGK1, PKM2, LDHA, proliferation such as IGF-2, TGFA, VEGFA, survival such as TERT, NANOG, OCT4 and cell migration-invasion such as ZEB1, ZEB2, SNAI2, MMP14, MMP9, AMF, MET, PTHrP. (Keith, et al Nat Rev Cancer 2012; 12:9-22).

In one embodiment, one or more signal-sensor polynucleotides may be administered to the cancer cell to investigate the destabilization of cancer, The selection of the sequence, dose or administrative route is optionally informed by diagnostic evaluation of the cell, tumor, tissue or organism including, but not limited to, expression profiling of the cancer, metabolic evaluation (hypoxic, acidotic), apoptotic vs. survival profiling, cell cycle vs. senescent profiling, immune sensitivities, and/or evaluation of stromal factors.

In one embodiment, the signal-sensor polynucleotides may encode either or both of the oncology related polypeptides, CITED4 and SHARP1. The signal-sensor polynucleotides are then administered where the administration of either or both results in the inhibition of the transcriptome of HIF-1 alpha in cancer cells. Suppression of HIF1-alpha gene regulated expression occurs upon administration with higher suppression when both polynucleotides are administered together. Reporter constructs such as luciferase under HIF1-alpha are used in the manner similar to the methods disclosed in van de Sluis et al, (J Clin Invest. 2010; 120(6):2119-2130). It is known that both CITED4 and SHARP1 expression results in decreased HIF1-alpha and concomitant reduction in HIF1-alpha regulated gene expression. Cell death and/or proliferation may also be evaluated in order to determine the effectiveness of the signal-sensor polynucleotide.

In another embodiment, additional experiments can be conducted using a cancer cell line where CITED4 and SHARP1 are themselves down regulated either under hypoxic conditions. A positive result would demonstrate that specifically targeting the metabolic profile (in this case hypoxic-adaptations of CITED4 and SHAPR1) with replacement of native proteins via signal-sensor polynucleotides can directly impact the transcriptome and survival advantage of cancer cells with this profile. Further, the data could show that the relative impact of signal-sensor polynucleotide vs. vehicle under hypoxic conditions was more significant for cancer cells than for normal cells. (i.e., the cancer cells have a disproportionate survival advantage based on their CITED4+SHARP1 down regulation) that makes them more sensitive to the replacement of this protein then a normal cell is to overproduction of it. It is understood that a cancer cell will likely be experiencing hypoxic conditions and that a normal cell under normoxic conditions might be able to tolerate CITED4 and SHARP1 over expression because the normal cell is not dependent on HIF1alpha transcriptome for survival advantage.

In one embodiment, in vivo experiments are performed according to the design of the in vitro experiments where the animal model is one evincing metastasis in the cancer setting because HIF-1alpha appears to confer the largest portion of its advantage in metastasis. Animals are administered the signal-sensor polynucleotide compared to no treatment or a control polynucleotide. Animal cells, tissues and/or organs are then evaluated for alterations in gene expression profiles or transcriptome levels.

Titration Between Cofactors

Experiments may be conducted in order to titrate the binding affinity between two cofactors. As used herein, the term “titrate” refers to a method whereby one or more factors are introduced systematically (such as at increasing levels or wherein the one or more factors are systematically modified) to a solution, scenario or series thereof in order to assess a property of interest. In this embodiment, the property of interest is the binding affinity between two cofactors. In one embodiment, constructs encoding the two cofactors are obtained and/or synthesized and a series of mutant constructs are prepared and/or synthesized. Mutant constructs encode cofactor mutants that may include truncated mutants (mutant proteins lacking one or more amino acids from either the N- or C-terminal domains), mutants with regional deletions [proteins wherein internal regions (comprising one or more amino acids) of the protein are absent], mutants with single amino acid substitutions (wherein a normally expressed amino acid is replaced with an alternative amino acid), mutants with one or more additional amino acids added internally or at either terminus, mutants with regional substitutions [proteins wherein internal regions (comprising one or more amino acids) of the protein are substituted with alternative regions (comprising one or more amino acids) and/or combinations of any of these. Mutant constructs are mutated randomly or subjected to targeted mutation based on existing knowledge of the molecular interactions necessary for binding between the two cofactors being investigated.

In some embodiments, a series of mutant proteins are designed such that the mutations follow a progressive pattern along the polypeptide chain. Such series may allow for a better understanding of a particular aspect or feature of the interaction between cofactors. A mutant series may include, for example, the production of a series of mutants, each with a single amino acid substitution, wherein each mutant has a different amino acid along it's polypeptide sequence mutated (e.g. alanine is substituted, thereby eliminating the influence of an amino acid side chain at each position). In another example, a series of mutants are designed such that the mutants in the series comprise truncations of increasing size. In another example, amino acids capable of being post-translationally modified (e.g. phosphorylated, acetylated, ubiquitinated, glycosylated, etc.) in a similar manner may be mutated along the polypeptide sequence in a series of mutants.

For titration experiments with mutant cofactors, a baseline affinity between the two cofactors is established by combining both cofactors under conditions favorable for binding and the binding affinity between the cofactors is assayed. Binding affinity may be assessed using any of a variety of methods known in the art. Such methods may include, but are not limited to Western blot analysis, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), fluorescence resonance energy transfer (FRET), fluorescence recovery after photobleaching (FRAP), fluorescence polarization technologies and/or surface plasmon resonance (SPR) based technologies. For titration, according to one method, a mutant series of one or both cofactors are combined with the two unmutated cofactors (to allow for binding competition between the wild type and mutated proteins). Changes in affinity between the two cofactors in the presence of increasing concentrations of different mutants are assessed and compared and/or plotted against the specific mutations present in the series of mutants that are competing for binding. Alternatively, mutant cofactors in a series are individually combined with a corresponding unmutated binding partner and assessed for binding affinity. Increasing concentrations of the wild type cofactor (corresponding to the mutant cofactor) are introduced and changes in binding between the mutant cofactors and the corresponding unmutated binding partner are assessed. Comparisons are made between the resulting binding curves and the binding curves of other mutants tested.

In some embodiments, titration of the binding affinity between two cofactors is assessed in the presence or absence of increasing concentrations of a third factor. Such a third factor may be an inhibitor or activator of binding between the two cofactors. A series of mutants, as described above, may be generated for a third factor and such a series may be used in titration experiments to assess the effect of mutations on binding between the two cofactors.

Information obtained from titration experiments may be used to design modified mRNA molecules to encode factors that modulate the interaction between cofactors.

In some embodiments, titration experiments are carried out wherein the binding affinity between HIF1 subunits (HIF1-alpha, HIF2-alpha and ARNT) and/or mutated HIF1 subunits and/or other proteins that interact with HIF1 is assessed. Titration experiments may utilize mutant series generated using constructs for one or more of HIF1-alpha, HIF2-alpha, ARNT and/or a third interacting factor. In some embodiments, a mutant series is generated for HIF1-alpha. HIF1-alpha and HIF2-alpha are hydroxylated by HIF hydroxylase enzymes under normal levels of oxygen in the cell, facilitating degradation and/or blocking transcriptional activity. Hydroxylation decreases as oxygen levels drop, allowing HIF1-alpha and/or HIF2-alpha to associate with their cofactor, ARNT leading to elevated expression of genes comprising HIF-response elements (HREs) (Keith, B. et al., HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer. 2011 Dec. 15; 12(1):9-22). In one embodiment, HIF1-alpha mutant series are generated wherein mutations in the series progressively eliminate one or more hydroxylation sites along the polypeptide chain (including, but not limited to proline 402, proline 564 and/or asparagine 803), thereby modulating stability and/or transcriptional activity in mutant versions of HIF1-alpha. In another embodiment, an alternative cofactor, HIF2-alpha is used to generate a mutant series. Such a mutant series may progressively eliminate one or more hydroxylation sites along the polypeptide chain (including, but not limited to proline 405, proline 531 and/or asparagine 847), thereby modulating stability and/or transcriptional activity in mutant versions of HIF2-alpha. In another embodiment, HIF1-alpha and/or HIF2-alpha mutant series are generated that progressively mutate regions necessary for interaction with ARNT, thereby creating mutants with altered abilities to bind ARNT and modulate HIF-dependent gene expression. In another embodiment, ARNT mutant series are generated that progressively mutate regions necessary for interactions with other HIF subunits, thereby creating mutants with altered abilities to bind HIF subunits and modulate HIF-dependent gene expression.

In some embodiments, mutant series are generated for Von Hippel-Landau tumor suppressor protein (pVHL). This protein binds hydroxylated HIF1-alpha and HIF2-alpha, facilitating their ubiquitination and degradation. In one embodiment, mutant series are generated that progressively mutate regions necessary for interaction with HIF1 subunits, thereby creating mutants with altered abilities to bind HIF1 subunits and modulate HIF-dependent gene expression.

Non-limiting examples of transcript and polypeptide sequences which may be used for the titration experiments are shown in Table 27 (transcript) and Table 28 (polypeptide).

Kits

The invention provides a variety of kits for conveniently and/or effectively carrying out methods of the present invention. Typically kits will comprise sufficient amounts and/or numbers of components to allow a user to perform multiple treatments of a subject(s) and/or to perform multiple experiments.

In one aspect, the present invention provides kits comprising the molecules (signal-sensor polynucleotides, primary constructs or mmRNA) of the invention. In one embodiment, the kit comprises one or more functional antibodies or function fragments thereof.

Said kits can be for oncology-related protein production, comprising a first signal-sensor polynucleotide, primary construct or mmRNA comprising a translatable region. The kit may further comprise packaging and instructions and/or a delivery agent to form a formulation composition. The delivery agent may comprise a saline, a buffered solution, a lipidoid or any delivery agent disclosed herein.

In one embodiment, the buffer solution may include sodium chloride, calcium chloride, phosphate and/or EDTA. In another embodiment, the buffer solution may include, but is not limited to, saline, saline with 2 mM calcium, 5% sucrose, 5% sucrose with 2 mM calcium, 5% Mannitol, 5% Mannitol with 2 mM calcium, Ringer's lactate, sodium chloride, sodium chloride with 2 mM calcium. In a further embodiment, the buffer solutions may be precipitated or it may be lyophilized. The amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations. The components may also be varied in order to increase the stability of modified RNA in the buffer solution over a period of time and/or under a variety of conditions. In one aspect, the present invention provides kits for oncology-related protein production, comprising: signal-sensor polynucleotide, primary construct or mmRNA comprising a translatable region, provided in an amount effective to produce a desired amount of an oncology-related protein encoded by the translatable region when introduced into a target cell; a second signal-sensor polynucleotide comprising an inhibitory nucleic acid, provided in an amount effective to substantially inhibit the innate immune response of the cell; and packaging and instructions.

In one aspect, the present invention provides kits for oncology-related protein production, comprising signal-sensor polynucleotide, primary construct or mmRNA comprising a translatable region, wherein the signal-sensor polynucleotide exhibits reduced degradation by a cellular nuclease, and packaging and instructions.

In one aspect, the present invention provides kits for oncology-related protein production, comprising signal-sensor polynucleotide, primary construct or mmRNA comprising a translatable region, wherein the polynucleotide exhibits reduced degradation by a cellular nuclease, and a mammalian cell suitable for translation of the translatable region of the first nucleic acid.

Devices

The present invention provides for devices which may incorporate signal-sensor polynucleotides, primary constructs or mmRNA that encode polypeptides of interest. These devices contain in a stable formulation the reagents to synthesize a signal-sensor polynucleotide in a formulation available to be immediately delivered to a subject in need thereof, such as a human patient.

In some embodiments the device is self-contained, and is optionally capable of wireless remote access to obtain instructions for synthesis and/or analysis of the generated signal-sensor polynucleotide, primary construct or mmRNA. The device is capable of mobile synthesis of at least one signal-sensor polynucleotide, primary construct or mmRNA and preferably an unlimited number of different signal-sensor polynucleotides, primary constructs or mmRNA. In certain embodiments, the device is capable of being transported by one or a small number of individuals. In other embodiments, the device is scaled to fit on a benchtop or desk. In other embodiments, the device is scaled to fit into a suitcase, backpack or similarly sized object. In another embodiment, the device may be a point of care or handheld device. In further embodiments, the device is scaled to fit into a vehicle, such as a car, truck or ambulance, or a military vehicle such as a tank or personnel carrier. The information necessary to generate a modified signal-sensor mRNA encoding oncology-related polypeptide of interest is present within a computer readable medium present in the device.

In one embodiment, a device may be used to assess levels of an oncology-related protein which has been administered in the form of signal-sensor polynucleotide, primary construct or mmRNA. The device may comprise a blood, urine or other biofluidic test.

In some embodiments, the device is capable of communication (e.g., wireless communication) with a database of nucleic acid and polypeptide sequences which may be signal-sensor nucleic acid and oncology-related polypeptide sequences. The device contains at least one sample block for insertion of one or more sample vessels. Such sample vessels are capable of accepting in liquid or other form any number of materials such as template DNA, nucleotides, enzymes, buffers, and other reagents. The sample vessels are also capable of being heated and cooled by contact with the sample block. The sample block is generally in communication with a device base with one or more electronic control units for the at least one sample block. The sample block preferably contains a heating module, such heating molecule capable of heating and/or cooling the sample vessels and contents thereof to temperatures between about −20 C and above +100 C. The device base is in communication with a voltage supply such as a battery or external voltage supply. The device also contains means for storing and distributing the materials for RNA synthesis.

Optionally, the sample block contains a module for separating the synthesized nucleic acids. Alternatively, the device contains a separation module operably linked to the sample block. Preferably the device contains a means for analysis of the synthesized nucleic acid. Such analysis includes sequence identity (demonstrated such as by hybridization), absence of non-desired sequences, measurement of integrity of synthesized mRNA (such has by microfluidic viscometry combined with spectrophotometry), and concentration and/or potency of modified RNA (such as by spectrophotometry).

In certain embodiments, the device is combined with a means for detection of pathogens present in a biological material obtained from a subject, e.g., the IBIS PLEX-ID system (Abbott, Abbott Park, Ill.) for microbial identification.

Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Pat. Nos. 4,886,499; 5,190,521; 5,328,483; 5,527,288; 4,270,537; 5,015,235; 5,141,496; and 5,417,662. Intradermal compositions may be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof. Jet injection devices which deliver liquid compositions to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S. Pat. Nos. 5,480,381; 5,599,302; 5,334,144; 5,993,412; 5,649,912; 5,569,189; 5,704,911; 5,383,851; 5,893,397; 5,466,220; 5,339,163; 5,312,335; 5,503,627; 5,064,413; 5,520,639; 4,596,556; 4,790,824; 4,941,880; 4,940,460; and PCT publications WO 97/37705 and WO 97/13537. Ballistic powder/particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable. Alternatively or additionally, conventional syringes may be used in the classical mantoux method of intradermal administration.

In some embodiments, the device may be a pump or comprise a catheter for administration of compounds or compositions of the invention across the blood brain barrier. Such devices include but are not limited to a pressurized olfactory delivery device, iontophoresis devices, multi-layered microfluidic devices, and the like. Such devices may be portable or stationary. They may be implantable or externally tethered to the body or combinations thereof.

Devices for administration may be employed to deliver the signal-sensor polynucleotides, primary constructs or mmRNA of the present invention according to single, multi- or split-dosing regimens taught herein. Such devices are described below.

Method and devices known in the art for multi-administration to cells, organs and tissues are contemplated for use in conjunction with the methods and compositions disclosed herein as embodiments of the present invention. These include, for example, those methods and devices having multiple needles, hybrid devices employing for example lumens or catheters as well as devices utilizing heat, electric current or radiation driven mechanisms.

According to the present invention, these multi-administration devices may be utilized to deliver the single, multi- or split doses contemplated herein.

A method for delivering therapeutic agents to a solid tissue has been described by Bahrami et al. and is taught for example in US Patent Publication 20110230839, the contents of which are incorporated herein by reference in their entirety. According to Bahrami, an array of needles is incorporated into a device which delivers a substantially equal amount of fluid at any location in said solid tissue along each needle's length.

A device for delivery of biological material across the biological tissue has been described by Kodgule et al. and is taught for example in US Patent Publication 20110172610, the contents of which are incorporated herein by reference in their entirety. According to Kodgule, multiple hollow micro-needles made of one or more metals and having outer diameters from about 200 microns to about 350 microns and lengths of at least 100 microns are incorporated into the device which delivers peptides, proteins, carbohydrates, nucleic acid molecules, lipids and other pharmaceutically active ingredients or combinations thereof.

A delivery probe for delivering a therapeutic agent to a tissue has been described by Gunday et al. and is taught for example in US Patent Publication 20110270184, the contents of which are incorporated herein by reference in their entirety. According to Gunday, multiple needles are incorporated into the device which moves the attached capsules between an activated position and an inactivated position to force the agent out of the capsules through the needles.

A multiple-injection medical apparatus has been described by Assaf and is taught for example in US Patent Publication 20110218497, the contents of which are incorporated herein by reference in their entirety. According to Assaf, multiple needles are incorporated into the device which has a chamber connected to one or more of said needles and a means for continuously refilling the chamber with the medical fluid after each injection.

In one embodiment, the signal-sensor polynucleotide, primary construct, or mmRNA is administered subcutaneously or intramuscularly via at least 3 needles to three different, optionally adjacent, sites simultaneously, or within a 60 minutes period (e.g., administration to 4, 5, 6, 7, 8, 9, or 10 sites simultaneously or within a 60 minute period). The split doses can be administered simultaneously to adjacent tissue using the devices described in U.S. Patent Publication Nos. 20110230839 and 20110218497, each of which is incorporated herein by reference.

An at least partially implantable system for injecting a substance into a patient's body, in particular a penis erection stimulation system has been described by Forsell and is taught for example in US Patent Publication 20110196198, the contents of which are incorporated herein by reference in their entirety. According to Forsell, multiple needles are incorporated into the device which is implanted along with one or more housings adjacent the patient's left and right corpora cavernosa. A reservoir and a pump are also implanted to supply drugs through the needles.

A method for the transdermal delivery of a therapeutic effective amount of iron has been described by Berenson and is taught for example in US Patent Publication 20100130910, the contents of which are incorporated herein by reference in their entirety. According to Berenson, multiple needles may be used to create multiple micro channels in stratum corneum to enhance transdermal delivery of the ionic iron on an iontophoretic patch.

A method for delivery of biological material across the biological tissue has been described by Kodgule et al and is taught for example in US Patent Publication 20110196308, the contents of which are incorporated herein by reference in their entirety. According to Kodgule, multiple biodegradable microneedles containing a therapeutic active ingredient are incorporated in a device which delivers proteins, carbohydrates, nucleic acid molecules, lipids and other pharmaceutically active ingredients or combinations thereof.

A transdermal patch comprising a botulinum toxin composition has been described by Donovan and is taught for example in US Patent Publication 20080220020, the contents of which are incorporated herein by reference in their entirety. According to Donovan, multiple needles are incorporated into the patch which delivers botulinum toxin under stratum corneum through said needles which project through the stratum corneum of the skin without rupturing a blood vessel.

A small, disposable drug reservoir, or patch pump, which can hold approximately 0.2 to 15 mL of liquid formulations can be placed on the skin and deliver the formulation continuously subcutaneously using a small bore needed (e.g., 26 to 34 gauge). As non-limiting examples, the patch pump may be 50 mm by 76 mm by 20 mm spring loaded having a 30 to 34 gauge needle (BD™ Microinfuser, Franklin Lakes N.J.), 41 mm by 62 mm by 17 mm with a 2 mL reservoir used for drug delivery such as insulin (OMNIPOD®, Insulet Corporation Bedford, Mass.), or 43-60 mm diameter, 10 mm thick with a 0.5 to 10 mL reservoir (PATCHPUMP®, SteadyMed Therapeutics, San Francisco, Calif.). Further, the patch pump may be battery powered and/or rechargeable.

A cryoprobe for administration of an active agent to a location of cryogenic treatment has been described by Toubia and is taught for example in US Patent Publication 20080140061, the contents of which are incorporated herein by reference in their entirety. According to Toubia, multiple needles are incorporated into the probe which receives the active agent into a chamber and administers the agent to the tissue.

A method for treating or preventing inflammation or promoting healthy joints has been described by Stock et al and is taught for example in US Patent Publication 20090155186, the contents of which are incorporated herein by reference in their entirety. According to Stock, multiple needles are incorporated in a device which administers compositions containing signal transduction modulator compounds.

A multi-site injection system has been described by Kimmell et al. and is taught for example in US Patent Publication 20100256594, the contents of which are incorporated herein by reference in their entirety. According to Kimmell, multiple needles are incorporated into a device which delivers a medication into a stratum corneum through the needles.

A method for delivering interferons to the intradermal compartment has been described by Dekker et al. and is taught for example in US Patent Publication 20050181033, the contents of which are incorporated herein by reference in their entirety. According to Dekker, multiple needles having an outlet with an exposed height between 0 and 1 mm are incorporated into a device which improves pharmaco*kinetics and bioavailability by delivering the substance at a depth between 0.3 mm and 2 mm.

A method for delivering genes, enzymes and biological agents to tissue cells has described by Desai and is taught for example in US Patent Publication 20030073908, the contents of which are incorporated herein by reference in their entirety. According to Desai, multiple needles are incorporated into a device which is inserted into a body and delivers a medication fluid through said needles.

A method for treating cardiac arrhythmias with fibroblast cells has been described by Lee et al and is taught for example in US Patent Publication 20040005295, the contents of which are incorporated herein by reference in their entirety. According to Lee, multiple needles are incorporated into the device which delivers fibroblast cells into the local region of the tissue.

A method using a magnetically controlled pump for treating a brain tumor has been described by Shachar et al. and is taught for example in U.S. Pat. No. 7,799,012 (method) and U.S. Pat. No. 7,799,016 (device), the contents of which are incorporated herein by reference in their entirety. According Shachar, multiple needles were incorporated into the pump which pushes a medicating agent through the needles at a controlled rate.

Methods of treating functional disorders of the bladder in mammalian females have been described by Versi et al. and are taught for example in U.S. Pat. No. 8,029,496, the contents of which are incorporated herein by reference in their entirety. According to Versi, an array of micro-needles is incorporated into a device which delivers a therapeutic agent through the needles directly into the trigone of the bladder.

A micro-needle transdermal transport device has been described by Angel et al and is taught for example in U.S. Pat. No. 7,364,568, the contents of which are incorporated herein by reference in their entirety. According to Angel, multiple needles are incorporated into the device which transports a substance into a body surface through the needles which are inserted into the surface from different directions. The micro-needle transdermal transport device may be a solid micro-needle system or a hollow micro-needle system. As a non-limiting example, the solid micro-needle system may have up to a 0.5 mg capacity, with 300-1500 solid micro-needles per cm2 about 150-700 μm tall coated with a drug. The micro-needles penetrate the stratum corneum and remain in the skin for short duration (e.g., 20 seconds to 15 minutes). In another example, the hollow micro-needle system has up to a 3 mL capacity to deliver liquid formulations using 15-20 microneedles per cm2 being approximately 950 μm tall. The micro-needles penetrate the skin to allow the liquid formulations to flow from the device into the skin. The hollow micro-needle system may be worn from 1 to 30 minutes depending on the formulation volume and viscosity.

A device for subcutaneous infusion has been described by Dalton et al and is taught for example in U.S. Pat. No. 7,150,726, the contents of which are incorporated herein by reference in their entirety. According to Dalton, multiple needles are incorporated into the device which delivers fluid through the needles into a subcutaneous tissue.

A device and a method for intradermal delivery of vaccines and gene therapeutic agents through microcannula have been described by Mikszta et al. and are taught for example in U.S. Pat. No. 7,473,247, the contents of which are incorporated herein by reference in their entirety. According to Mitszta, at least one hollow micro-needle is incorporated into the device which delivers the vaccines to the subject's skin to a depth of between 0.025 mm and 2 mm.

A method of delivering insulin has been described by Pettis et al and is taught for example in U.S. Pat. No. 7,722,595, the contents of which are incorporated herein by reference in their entirety. According to Pettis, two needles are incorporated into a device wherein both needles insert essentially simultaneously into the skin with the first at a depth of less than 2.5 mm to deliver insulin to intradermal compartment and the second at a depth of greater than 2.5 mm and less than 5.0 mm to deliver insulin to subcutaneous compartment.

Cutaneous injection delivery under suction has been described by Kochamba et al. and is taught for example in U.S. Pat. No. 6,896,666, the contents of which are incorporated herein by reference in their entirety. According to Kochamba, multiple needles in relative adjacency with each other are incorporated into a device which injects a fluid below the cutaneous layer.

A device for withdrawing or delivering a substance through the skin has been described by Down et al and is taught for example in U.S. Pat. No. 6,607,513, the contents of which are incorporated herein by reference in their entirety. According to Down, multiple skin penetrating members which are incorporated into the device have lengths of about 100 microns to about 2000 microns and are about 30 to 50 gauge.

A device for delivering a substance to the skin has been described by Palmer et al and is taught for example in U.S. Pat. No. 6,537,242, the contents of which are incorporated herein by reference in their entirety. According to Palmer, an array of micro-needles is incorporated into the device which uses a stretching assembly to enhance the contact of the needles with the skin and provides a more uniform delivery of the substance.

A perfusion device for localized drug delivery has been described by Zamoyski and is taught for example in U.S. Pat. No. 6,468,247, the contents of which are incorporated herein by reference in their entirety. According to Zamoyski, multiple hypodermic needles are incorporated into the device which injects the contents of the hypodermics into a tissue as said hypodermics are being retracted.

A method for enhanced transport of drugs and biological molecules across tissue by improving the interaction between micro-needles and human skin has been described by Prausnitz et al. and is taught for example in U.S. Pat. No. 6,743,211, the contents of which are incorporated herein by reference in their entirety. According to Prausnitz, multiple micro-needles are incorporated into a device which is able to present a more rigid and less deformable surface to which the micro-needles are applied.

A device for intraorgan administration of medicinal agents has been described by Ting et al and is taught for example in U.S. Pat. No. 6,077,251, the contents of which are incorporated herein by reference in their entirety. According to Ting, multiple needles having side openings for enhanced administration are incorporated into a device which by extending and retracting said needles from and into the needle chamber forces a medicinal agent from a reservoir into said needles and injects said medicinal agent into a target organ.

A multiple needle holder and a subcutaneous multiple channel infusion port has been described by Brown and is taught for example in U.S. Pat. No. 4,695,273, the contents of which are incorporated herein by reference in their entirety. According to Brown, multiple needles on the needle holder are inserted through the septum of the infusion port and communicate with isolated chambers in said infusion port.

A dual hypodermic syringe has been described by Horn and is taught for example in U.S. Pat. No. 3,552,394, the contents of which are incorporated herein by reference in their entirety. According to Horn, two needles incorporated into the device are spaced apart less than 68 mm and may be of different styles and lengths, thus enabling injections to be made to different depths.

A syringe with multiple needles and multiple fluid compartments has been described by Hershberg and is taught for example in U.S. Pat. No. 3,572,336, the contents of which are incorporated herein by reference in their entirety. According to Hershberg, multiple needles are incorporated into the syringe which has multiple fluid compartments and is capable of simultaneously administering incompatible drugs which are not able to be mixed for one injection.

A surgical instrument for intradermal injection of fluids has been described by Eliscu et al. and is taught for example in U.S. Pat. No. 2,588,623, the contents of which are incorporated herein by reference in their entirety. According to Eliscu, multiple needles are incorporated into the instrument which injects fluids intradermally with a wider disperse.

An apparatus for simultaneous delivery of a substance to multiple breast milk ducts has been described by Hung and is taught for example in EP 1818017, the contents of which are incorporated herein by reference in their entirety. According to Hung, multiple lumens are incorporated into the device which inserts though the orifices of the ductal networks and delivers a fluid to the ductal networks.

A catheter for introduction of medications to the tissue of a heart or other organs has been described by Tkebuchava and is taught for example in WO2006138109, the contents of which are incorporated herein by reference in their entirety. According to Tkebuchava, two curved needles are incorporated which enter the organ wall in a flattened trajectory.

Devices for delivering medical agents have been described by Mckay et al. and are taught for example in WO2006118804, the content of which are incorporated herein by reference in their entirety. According to Mckay, multiple needles with multiple orifices on each needle are incorporated into the devices to facilitate regional delivery to a tissue, such as the interior disc space of a spinal disc.

A method for directly delivering an immunomodulatory substance into an intradermal space within a mammalian skin has been described by Pettis and is taught for example in WO2004020014, the contents of which are incorporated herein by reference in their entirety. According to Pettis, multiple needles are incorporated into a device which delivers the substance through the needles to a depth between 0.3 mm and 2 mm.

Methods and devices for administration of substances into at least two compartments in skin for systemic absorption and improved pharmaco*kinetics have been described by Pettis et al. and are taught for example in WO2003094995, the contents of which are incorporated herein by reference in their entirety. According to Pettis, multiple needles having lengths between about 300 μm and about 5 mm are incorporated into a device which delivers to intradermal and subcutaneous tissue compartments simultaneously.

A drug delivery device with needles and a roller has been described by Zimmerman et al. and is taught for example in WO2012006259, the contents of which are incorporated herein by reference in their entirety. According to Zimmerman, multiple hollow needles positioned in a roller are incorporated into the device which delivers the content in a reservoir through the needles as the roller rotates.

Methods and Devices Utilizing Catheters and/or Lumens

Methods and devices using catheters and lumens may be employed to administer the mmRNA of the present invention on a single, multi- or split dosing schedule. Such methods and devices are described below.

A catheter-based delivery of skeletal myoblasts to the myocardium of damaged hearts has been described by Jacoby et al and is taught for example in US Patent Publication 20060263338, the contents of which are incorporated herein by reference in their entirety. According to Jacoby, multiple needles are incorporated into the device at least part of which is inserted into a blood vessel and delivers the cell composition through the needles into the localized region of the subject's heart.

An apparatus for treating asthma using neurotoxin has been described by Deem et al and is taught for example in US Patent Publication 20060225742, the contents of which are incorporated herein by reference in their entirety. According to Deem, multiple needles are incorporated into the device which delivers neurotoxin through the needles into the bronchial tissue.

A method for administering multiple-component therapies has been described by Nayak and is taught for example in U.S. Pat. No. 7,699,803, the contents of which are incorporated herein by reference in their entirety. According to Nayak, multiple injection cannulas may be incorporated into a device wherein depth slots may be included for controlling the depth at which the therapeutic substance is delivered within the tissue.

A surgical device for ablating a channel and delivering at least one therapeutic agent into a desired region of the tissue has been described by McIntyre et al and is taught for example in U.S. Pat. No. 8,012,096, the contents of which are incorporated herein by reference in their entirety. According to McIntyre, multiple needles are incorporated into the device which dispenses a therapeutic agent into a region of tissue surrounding the channel and is particularly well suited for transmyocardial revascularization operations.

Methods of treating functional disorders of the bladder in mammalian females have been described by Versi et al and are taught for example in U.S. Pat. No. 8,029,496, the contents of which are incorporated herein by reference in their entirety. According to Versi, an array of micro-needles is incorporated into a device which delivers a therapeutic agent through the needles directly into the trigone of the bladder.

A device and a method for delivering fluid into a flexible biological barrier have been described by Yeshurun et al. and are taught for example in U.S. Pat. No. 7,998,119 (device) and U.S. Pat. No. 8,007,466 (method), the contents of which are incorporated herein by reference in their entirety. According to Yeshurun, the micro-needles on the device penetrate and extend into the flexible biological barrier and fluid is injected through the bore of the hollow micro-needles.

A method for epicardially injecting a substance into an area of tissue of a heart having an epicardial surface and disposed within a torso has been described by Bonner et al and is taught for example in U.S. Pat. No. 7,628,780, the contents of which are incorporated herein by reference in their entirety. According to Bonner, the devices have elongate shafts and distal injection heads for driving needles into tissue and injecting medical agents into the tissue through the needles.

A device for sealing a puncture has been described by Nielsen et al and is taught for example in U.S. Pat. No. 7,972,358, the contents of which are incorporated herein by reference in their entirety. According to Nielsen, multiple needles are incorporated into the device which delivers a closure agent into the tissue surrounding the puncture tract.

A method for myogenesis and angiogenesis has been described by Chiu et al. and is taught for example in U.S. Pat. No. 6,551,338, the contents of which are incorporated herein by reference in their entirety. According to Chiu, 5 to 15 needles having a maximum diameter of at least 1.25 mm and a length effective to provide a puncture depth of 6 to 20 mm are incorporated into a device which inserts into proximity with a myocardium and supplies an exogeneous angiogenic or myogenic factor to said myocardium through the conduits which are in at least some of said needles.

A method for the treatment of prostate tissue has been described by Bolmsj et al. and is taught for example in U.S. Pat. No. 6,524,270, the contents of which are incorporated herein by reference in their entirety. According to Bolmsj, a device comprising a catheter which is inserted through the urethra has at least one hollow tip extendible into the surrounding prostate tissue. An astringent and analgesic medicine is administered through said tip into said prostate tissue.

A method for infusing fluids to an intraosseous site has been described by Findlay et al. and is taught for example in U.S. Pat. No. 6,761,726, the contents of which are incorporated herein by reference in their entirety. According to Findlay, multiple needles are incorporated into a device which is capable of penetrating a hard shell of material covered by a layer of soft material and delivers a fluid at a predetermined distance below said hard shell of material.

A device for injecting medications into a vessel wall has been described by Vigil et al. and is taught for example in U.S. Pat. No. 5,713,863, the contents of which are incorporated herein by reference in their entirety. According to Vigil, multiple injectors are mounted on each of the flexible tubes in the device which introduces a medication fluid through a multi-lumen catheter, into said flexible tubes and out of said injectors for infusion into the vessel wall.

A catheter for delivering therapeutic and/or diagnostic agents to the tissue surrounding a bodily passageway has been described by Faxon et al. and is taught for example in U.S. Pat. No. 5,464,395, the contents of which are incorporated herein by reference in their entirety. According to Faxon, at least one needle cannula is incorporated into the catheter which delivers the desired agents to the tissue through said needles which project outboard of the catheter.

Balloon catheters for delivering therapeutic agents have been described by Orr and are taught for example in WO2010024871, the contents of which are incorporated herein by reference in their entirety. According to Orr, multiple needles are incorporated into the devices which deliver the therapeutic agents to different depths within the tissue.

Methods and Devices Utilizing Electrical Current

Methods and devices utilizing electric current may be employed to deliver the mmRNA of the present invention according to the single, multi- or split dosing regimens taught herein. Such methods and devices are described below.

An electro collagen induction therapy device has been described by Marquez and is taught for example in US Patent Publication 20090137945, the contents of which are incorporated herein by reference in their entirety. According to Marquez, multiple needles are incorporated into the device which repeatedly pierce the skin and draw in the skin a portion of the substance which is applied to the skin first.

An electrokinetic system has been described by Etheredge et al. and is taught for example in US Patent Publication 20070185432, the contents of which are incorporated herein by reference in their entirety. According to Etheredge, micro-needles are incorporated into a device which drives by an electrical current the medication through the needles into the targeted treatment site.

An iontophoresis device has been described by Matsumura et al. and is taught for example in U.S. Pat. No. 7,437,189, the contents of which are incorporated herein by reference in their entirety. According to Matsumura, multiple needles are incorporated into the device which is capable of delivering ionizable drug into a living body at higher speed or with higher efficiency.

Intradermal delivery of biologically active agents by needle-free injection and electroporation has been described by Hoffmann et al and is taught for example in U.S. Pat. No. 7,171,264, the contents of which are incorporated herein by reference in their entirety. According to Hoffmann, one or more needle-free injectors are incorporated into an electroporation device and the combination of needle-free injection and electroporation is sufficient to introduce the agent into cells in skin, muscle or mucosa.

A method for electropermeabilization-mediated intracellular delivery has been described by Lundkvist et al. and is taught for example in U.S. Pat. No. 6,625,486, the contents of which are incorporated herein by reference in their entirety. According to Lundkvist, a pair of needle electrodes is incorporated into a catheter. Said catheter is positioned into a body lumen followed by extending said needle electrodes to penetrate into the tissue surrounding said lumen. Then the device introduces an agent through at least one of said needle electrodes and applies electric field by said pair of needle electrodes to allow said agent pass through the cell membranes into the cells at the treatment site.

A delivery system for transdermal immunization has been described by Levin et al. and is taught for example in WO2006003659, the contents of which are incorporated herein by reference in their entirety. According to Levin, multiple electrodes are incorporated into the device which applies electrical energy between the electrodes to generate micro channels in the skin to facilitate transdermal delivery.

A method for delivering RF energy into skin has been described by Schomacker and is taught for example in WO2011163264, the contents of which are incorporated herein by reference in their entirety. According to Schomacker, multiple needles are incorporated into a device which applies vacuum to draw skin into contact with a plate so that needles insert into skin through the holes on the plate and deliver RF energy.

At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. For example, the term “C1-6 alkyl” is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl.

About: As used herein, the term “about” means +/−10% of the recited value.

Administered in combination: As used herein, the term “administered in combination” or “combined administration” means that two or more agents are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient. In some embodiments, they are administered within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced sufficiently closely together such that a combinatorial (e.g., a synergistic) effect is achieved.

Animal: As used herein, the term “animal” refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans at any stage of development. In some embodiments, “animal” refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.

Antigens of interest or desired antigens: As used herein, the terms “antigens of interest” or “desired antigens” include those proteins and other biomolecules provided herein that are immunospecifically bound by the antibodies and fragments, mutants, variants, and alterations thereof described herein. Examples of antigens of interest include, but are not limited to, insulin, insulin-like growth factor, hGH, tPA, cytokines, such as interleukins (IL), e.g., IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, interferon (IFN) alpha, IFN beta, IFN gamma, IFN omega or IFN tau, tumor necrosis factor (TNF), such as TNF alpha and TNF beta, TNF gamma, TRAIL; G-CSF, GM-CSF, M-CSF, MCP-1 and VEGF.

Approximately: As used herein, the term “approximately” or “about,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term “approximately” or “about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).

Associated with: As used herein, the terms “associated with,” “conjugated,” “linked,” “attached,” and “tethered,” when used with respect to two or more moieties, means that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serves as a linking agent, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g., physiological conditions. An “association” need not be strictly through direct covalent chemical bonding. It may also suggest ionic or hydrogen bonding or a hybridization based connectivity sufficiently stable such that the “associated” entities remain physically associated.

Bifunctional: As used herein, the term “bifunctional” refers to any substance, molecule or moiety which is capable of or maintains at least two functions. The functions may effect the same outcome or a different outcome. The structure that produces the function may be the same or different. For example, bifunctional modified RNAs of the present invention may encode a cytotoxic peptide (a first function) while those nucleosides which comprise the encoding RNA are, in and of themselves, cytotoxic (second function). In this example, delivery of the bifunctional modified RNA to a cancer cell would produce not only a peptide or protein molecule which may ameliorate or treat the cancer but would also deliver a cytotoxic payload of nucleosides to the cell should degradation, instead of translation of the modified RNA, occur.

Biocompatible: As used herein, the term “biocompatible” means compatible with living cells, tissues, organs or systems posing little to no risk of injury, toxicity or rejection by the immune system.

Biodegradable: As used herein, the term “biodegradable” means capable of being broken down into innocuous products by the action of living things.

Biologically active: As used herein, the phrase “biologically active” refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active. In particular embodiments, signal-sensor polynucleotide, primary construct or mmRNA of the present invention may be considered biologically active if even a portion of the signal-sensor polynucleotide, primary construct or mmRNA is biologically active or mimics an activity considered biologically relevant.

Cancer: As used herein, the term “cancer” in a subject refers to the presence of cells possessing characteristics, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Often, cancer cells will be in the form of a tumor, but such cells may exist alone within a subject, or may circulate in the blood stream as independent cells, such as leukemic cells.

Cell growth: As used herein, the term “cell growth” is principally associated with growth in cell numbers, which occurs by means of cell reproduction (i.e. proliferation) when the rate of the latter is greater than the rate of cell death (e.g. by apoptosis or necrosis).

Chemical terms: The following provides the definition of various chemical terms from “acyl” to “thiol.”

The term “acyl,” as used herein, represents a hydrogen or an alkyl group (e.g., a haloalkyl group), as defined herein, that is attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde group), acetyl, propionyl, butanoyl and the like. Exemplary unsubstituted acyl groups include from 1 to 7, from 1 to 11, or from 1 to 21 carbons. In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein.

The term “acylamino,” as used herein, represents an acyl group, as defined herein, attached to the parent molecular group though an amino group, as defined herein (i.e., —N(RN1)—C(O)—R, where R is H or an optionally substituted C1-6, C1-10, or C1-20 alkyl group and RN1 is as defined herein). Exemplary unsubstituted acylamino groups include from 1 to 41 carbons (e.g., from 1 to 7, from 1 to 13, from 1 to 21, from 2 to 7, from 2 to 13, from 2 to 21, or from 2 to 41 carbons). In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein, and/or the amino group is —NH2 or —NHRN1, wherein RN1 is, independently, OH, NO2, NH2, NRN22, SO2ORN2, SO2RN2, SORN2, alkyl, or aryl, and each RN2 can be H, alkyl, or aryl.

The term “acyloxy,” as used herein, represents an acyl group, as defined herein, attached to the parent molecular group though an oxygen atom (i.e., —O—C(O)—R, where R is H or an optionally substituted C1-6, C1-10, or C1-20 alkyl group). Exemplary unsubstituted acyloxy groups include from 1 to 21 carbons (e.g., from 1 to 7 or from 1 to 11 carbons). In some embodiments, the alkyl group is further substituted with 1, 2, 3, or 4 substituents as described herein, and/or the amino group is —NH2, or —NHRN1, wherein is, independently, OH, NO2, NH2, NRN22, SO2ORN2, SO2RN2, SORN2, alkyl, or aryl, and each RN2 can be H, alkyl, or aryl.

The term “alkaryl,” as used herein, represents an aryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkaryl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-6 alk-C6-10 aryl, C1-10 alk-C6-10 aryl, or C1-20 alk-C6-10 aryl). In some embodiments, the alkylene and the aryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective groups. Other groups preceded by the prefix “alk-” are defined in the same manner, where “alk” refers to a C1-6 alkylene, unless otherwise noted, and the attached chemical structure is as defined herein.

The term “alkcycloalkyl” represents a cycloalkyl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein (e.g., an alkylene group of from 1 to 4, from 1 to 6, from 1 to 10, or form 1 to 20 carbons). In some embodiments, the alkylene and the cycloalkyl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group.

The term “alkenyl,” as used herein, represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 20 carbons (e.g., from 2 to 6 or from 2 to 10 carbons) containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, and the like. Alkenyls include both cis and trans isomers. Alkenyl groups may be optionally substituted with 1, 2, 3, or 4 substituent groups that are selected, independently, from amino, aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.

The term “alkenyloxy” represents a chemical substituent of formula —OR, where R is a C2-20 alkenyl group (e.g., C2-6 or C2-10 alkenyl), unless otherwise specified. Exemplary alkenyloxy groups include ethenyloxy, propenyloxy, and the like. In some embodiments, the alkenyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein (e.g., a hydroxy group).

The term “alkheteroaryl” refers to a heteroaryl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkheteroaryl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to 18, from 2 to 17, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as C1-6 alk-C1-12 heteroaryl, C1-10 alk-C1-12 heteroaryl, or C1-20 alk-C1-12 heteroaryl). In some embodiments, the alkylene and the heteroaryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group. Alkheteroaryl groups are a subset of alkheterocyclyl groups.

The term “alkheterocyclyl” represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an alkylene group, as defined herein. Exemplary unsubstituted alkheterocyclyl groups are from 2 to 32 carbons (e.g., from 2 to 22, from 2 to 18, from 2 to 17, from 2 to 16, from 3 to 15, from 2 to 14, from 2 to 13, or from 2 to 12 carbons, such as C1-6 alk-C1-12 heterocyclyl, C1-10 alk-C1-12 heterocyclyl, or alk-C1-12 heterocyclyl). In some embodiments, the alkylene and the heterocyclyl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group.

The term “alkoxy” represents a chemical substituent of formula —OR, where R is a C1-20 alkyl group (e.g., C1-6 or C1-10 alkyl), unless otherwise specified. Exemplary alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein (e.g., hydroxy or alkoxy).

The term “alkoxyalkoxy” represents an alkoxy group that is substituted with an alkoxy group. Exemplary unsubstituted alkoxyalkoxy groups include between 2 to 40 carbons (e.g., from 2 to 12 or from 2 to 20 carbons, such as C1-6 alkoxy-C1-6 alkoxy, C1-10 alkoxy-C1-10 alkoxy, or C1-20 alkoxy-C1-20 alkoxy). In some embodiments, the each alkoxy group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein.

The term “alkoxyalkyl” represents an alkyl group that is substituted with an alkoxy group. Exemplary unsubstituted alkoxyalkyl groups include between 2 to 40 carbons (e.g., from 2 to 12 or from 2 to 20 carbons, such as C1-6 alkoxy-C1-6 alkyl, C1-10 alkoxy-C1-10 alkyl, or C1-20 alkoxy-C1-20 alkyl). In some embodiments, the alkyl and the alkoxy each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective group.

The term “alkoxycarbonyl,” as used herein, represents an alkoxy, as defined herein, attached to the parent molecular group through a carbonyl atom (e.g., —C(O)—OR, where R is H or an optionally substituted C1-6, C1-10, or C1-20 alkyl group). Exemplary unsubstituted alkoxycarbonyl include from 1 to 21 carbons (e.g., from 1 to 11 or from 1 to 7 carbons). In some embodiments, the alkoxy group is further substituted with 1, 2, 3, or 4 substituents as described herein.

The term “alkoxycarbonylalkoxy,” as used herein, represents an alkoxy group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., —O-alkyl-C(O)—OR, where R is an optionally substituted C1-6, C1-10, or C1-20 alkyl group). Exemplary unsubstituted alkoxycarbonylalkoxy include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as C1-6 alkoxycarbonyl-C1-6 alkoxy, C1-10 alkoxycarbonyl-C1-10 alkoxy, or C1-20 alkoxycarbonyl-C1-20 alkoxy). In some embodiments, each alkoxy group is further independently substituted with 1, 2, 3, or 4 substituents, as described herein (e.g., a hydroxy group).

The term “alkoxycarbonylalkyl,” as used herein, represents an alkyl group, as defined herein, that is substituted with an alkoxycarbonyl group, as defined herein (e.g., -alkyl-C(O)—OR, where R is an optionally substituted C1-20, C1-10, or C1-6 alkyl group). Exemplary unsubstituted alkoxycarbonylalkyl include from 3 to 41 carbons (e.g., from 3 to 10, from 3 to 13, from 3 to 17, from 3 to 21, or from 3 to 31 carbons, such as C1-6 alkoxycarbonyl-C1-6 alkyl, C1-10 alkoxycarbonyl-C1-10 alkyl, or C1-20 alkoxycarbonyl-C1-20 alkyl). In some embodiments, each alkyl and alkoxy group is further independently substituted with 1, 2, 3, or 4 substituents as described herein (e.g., a hydroxy group).

The term “alkyl,” as used herein, is inclusive of both straight chain and branched chain saturated groups from 1 to 20 carbons (e.g., from 1 to 10 or from 1 to 6), unless otherwise specified. Alkyl groups are exemplified by methyl, ethyl, n- and isopropyl, n-, sec-, iso- and tert-butyl, neopentyl, and the like, and may be optionally substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four substituents independently selected from the group consisting of: (1) C1-6 alkoxy; (2) C1-6 alkylsulfinyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e., —NH2) or a substituted amino (i.e., —N(RN1)2, where RN1 is as defined for amino); (4) C6-10 aryl-C1-6 alkoxy; (5) azido; (6) halo; (7) (C2-9 heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11) C1-7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) —CO2RA′, where RA′ is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) C6-10 aryl, (d) hydrogen, (e) C1-6 alk-C6-10 aryl, (f) amino-C1-20 alkyl, (g) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (15) —C(O)NRB′RC′, where each of RB′ and RC′ is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (16) —SO2RD′, where RD′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) C1-6 alk-C6-10 aryl, and (d) hydroxy; (17) —SO2NRE′RF′, where each of RE′ and RF′ is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl and (d) C1-6 alk-C6-10 aryl; (18) —C(O)RG′, where RG′ is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) C6-10 aryl, (d) hydrogen, (e) C1-6 alk-C6-10 aryl, (f) amino-C1-20 alkyl, (g) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (19) —NRH′C(O)RI′, wherein RH′ is selected from the group consisting of (a1) hydrogen and (b1) C1-6 alkyl, and RI′ is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) C6-10 aryl, (d2) hydrogen, (e2) C1-6 alk-C6-10 aryl, (f2) amino-C1-20 alkyl, (g2) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h2) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (20) —NRJ′C(O)ORK′, wherein RJ′ is selected from the group consisting of (a1) hydrogen and (b1) C1-6 alkyl, and RK′ is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) C6-10 aryl, (d2) hydrogen, (e2) alk-C6-10 aryl, (f2) amino-C1-20 alkyl, (g2) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h2) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a C1-alkaryl can be further substituted with an oxo group to afford the respective aryloyl substituent.

The term “alkylene” and the prefix “alk-,” as used herein, represent a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like. The term “Cx-y alkylene” and the prefix “Cx-y alk-” represent alkylene groups having between x and y carbons. Exemplary values for x are 1, 2, 3, 4, 5, and 6, and exemplary values for y are 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 (e.g., C1-6, C1-10, C2-20, C2-6, C2-10, or C2-20 alkylene). In some embodiments, the alkylene can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for an alkyl group.

The term “alkylsulfinyl,” as used herein, represents an alkyl group attached to the parent molecular group through an —S(O)— group. Exemplary unsubstituted alkylsulfinyl groups are from 1 to 6, from 1 to 10, or from 1 to 20 carbons. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein.

The term “alkylsulfinylalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by an alkylsulfinyl group. Exemplary unsubstituted alkylsulfinylalkyl groups are from 2 to 12, from 2 to 20, or from 2 to 40 carbons. In some embodiments, each alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein.

The term “alkynyl,” as used herein, represents monovalent straight or branched chain groups from 2 to 20 carbon atoms (e.g., from 2 to 4, from 2 to 6, or from 2 to 10 carbons) containing a carbon-carbon triple bond and is exemplified by ethynyl, 1-propynyl, and the like. Alkynyl groups may be optionally substituted with 1, 2, 3, or 4 substituent groups that are selected, independently, from aryl, cycloalkyl, or heterocyclyl (e.g., heteroaryl), as defined herein, or any of the exemplary alkyl substituent groups described herein.

The term “alkynyloxy” represents a chemical substituent of formula —OR, where R is a C2-20 alkynyl group (e.g., C2-6 or C2-10 alkynyl), unless otherwise specified. Exemplary alkynyloxy groups include ethynyloxy, propynyloxy, and the like. In some embodiments, the alkynyl group can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein (e.g., a hydroxy group).

The term “amidine,” as used herein, represents a —C(═NH)NH2 group.

The term “amino,” as used herein, represents —N(RN1)2, wherein each RN1 is, independently, H, OH, NO2, N(RN2)2, SO2ORN2, SO2RN2, SORN2, an A-protecting group, alkyl, alkenyl, alkynyl, alkoxy, aryl, alkaryl, cycloalkyl, alkcycloalkyl, carboxyalkyl, sulfoalkyl, heterocyclyl (e.g., heteroaryl), or alkheterocyclyl (e.g., alkheteroaryl), wherein each of these recited RN1 groups can be optionally substituted, as defined herein for each group; or two RN1 combine to form a heterocyclyl or an N-protecting group, and wherein each RN2 is, independently, H, alkyl, or aryl. The amino groups of the invention can be an unsubstituted amino (i.e., —NH2) or a substituted amino (i.e., —N(RN1)2). In a preferred embodiment, amino is —NH2 or —NHRN1, wherein RN1 is, independently, OH, NO2, NH2, NRN22, SO2ORN2, SO2RN2, SORN2, alkyl, carboxyalkyl, sulfoalkyl, or aryl, and each RN2 can be H, C1-20 alkyl (e.g., C1-6 alkyl), or C6-10 aryl.

The term “amino acid,” as described herein, refers to a molecule having a side chain, an amino group, and an acid group (e.g., a carboxy group of —CO2H or a sulfo group of —SO3H), wherein the amino acid is attached to the parent molecular group by the side chain, amino group, or acid group (e.g., the side chain). In some embodiments, the amino acid is attached to the parent molecular group by a carbonyl group, where the side chain or amino group is attached to the carbonyl group. Exemplary side chains include an optionally substituted alkyl, aryl, heterocyclyl, alkaryl, alkheterocyclyl, aminoalkyl, carbamoylalkyl, and carboxyalkyl. Exemplary amino acids include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, hydroxynorvaline, isoleucine, leucine, lysine, methionine, norvaline, ornithine, phenylalanine, proline, pyrrolysine, selenocysteine, serine, taurine, threonine, tryptophan, tyrosine, and valine. Amino acid groups may be optionally substituted with one, two, three, or, in the case of amino acid groups of two carbons or more, four substituents independently selected from the group consisting of: (1) C1-6 alkoxy; (2) C1-6 alkylsulfinyl; (3) amino, as defined herein (e.g., unsubstituted amino (i.e., —NH2) or a substituted amino (i.e., —N(RN1)2, where RN1 is as defined for amino); (4) C6-10 aryl-C1-6 alkoxy; (5) azido; (6) halo; (7) (C2-9 heterocyclyl)oxy; (8) hydroxy; (9) nitro; (10) oxo (e.g., carboxyaldehyde or acyl); (11) C1-7 spirocyclyl; (12) thioalkoxy; (13) thiol; (14) —CO2RA′, where RA′ is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) C6-10 aryl, (d) hydrogen, (e) C1-6 alk-C6-10 aryl, (f) amino-C1-20 alkyl, (g) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (15) —C(O)NRB′RC′, where each of RB′ and RC′ is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (16) —SO2RD′, where RD′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) C1-6 alk-C6-10 aryl, and (d) hydroxy; (17) —SO2NRE′RF′, where each of RE′ and RF′ is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl and (d) C1-6 alk-C6-10 aryl; (18) —C(O)RG′, where RG′ is selected from the group consisting of (a) C1-20 alkyl (e.g., C1-6 alkyl), (b) C2-20 alkenyl (e.g., C2-6 alkenyl), (c) C6-10 aryl, (d) hydrogen, (e) C1-6 alk-C6-10 aryl, (f) amino-C1-20 alkyl, (g) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (19) —NRH′C(O)RI′, wherein RH′ is selected from the group consisting of (a1) hydrogen and (b1) C1-6 alkyl, and RI′ is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) C6-10 aryl, (d2) hydrogen, (e2) C1-6 alk-C6-10 aryl, (f2) amino-C1-20 alkyl, (g2) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h2) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; (20) —NRJ′C(O)ORK′, wherein RJ′ is selected from the group consisting of (a1) hydrogen and (b1) alkyl, and RK′ is selected from the group consisting of (a2) C1-20 alkyl (e.g., C1-6 alkyl), (b2) C2-20 alkenyl (e.g., C2-6 alkenyl), (c2) C6-10 aryl, (d2) hydrogen, (e2) C1-6 alk-C6-10 aryl, (f2) amino-C1-20 alkyl, (g2) polyethylene glycol of —(CH2)s2(OCH2CH2)s1(CH2)s3OR′, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and R′ is H or C1-20 alkyl, and (h2) amino-polyethylene glycol of —NRN1(CH2)s2(CH2CH2O)s1(CH2)s3NRN1, wherein s1 is an integer from 1 to 10 (e.g., from 1 to 6 or from 1 to 4), each of s2 and s3, independently, is an integer from 0 to 10 (e.g., from 0 to 4, from 0 to 6, from 1 to 4, from 1 to 6, or from 1 to 10), and each RN1 is, independently, hydrogen or optionally substituted C1-6 alkyl; and (21) amidine. In some embodiments, each of these groups can be further substituted as described herein.

The term “aminoalkoxy,” as used herein, represents an alkoxy group, as defined herein, substituted by an amino group, as defined herein. The alkyl and amino each can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., CO2RA′, where RA′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) hydrogen, and (d) C1-6 alk-C6-10 aryl, e.g., carboxy).

The term “aminoalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by an amino group, as defined herein. The alkyl and amino each can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the respective group (e.g., CO2RA′, where RA′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) hydrogen, and (d) C1-6 alk-C6-10 aryl, e.g., carboxy).

The term “aryl,” as used herein, represents a mono-, bicyclic, or multicyclic carbocyclic ring system having one or two aromatic rings and is exemplified by phenyl, naphthyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, anthracenyl, phenanthrenyl, fluorenyl, indanyl, indenyl, and the like, and may be optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from the group consisting of: (1) C1-7 acyl (e.g., carboxyaldehyde); (2) C1-20 alkyl (e.g., C1-6 alkyl, C1-6 alkoxy-C1-6 alkyl, C1-6 alkylsulfinyl-C1-6 alkyl, amino-C1-6 alkyl, azido-C1-6 alkyl, (carboxyaldehyde)-C1-6 alkyl, halo-C1-6 alkyl (e.g., perfluoroalkyl), hydroxy-C1-6 alkyl, nitro-C1-6 alkyl, or C1-6 thioalkoxy-C1-6 alkyl); (3) C1-20 alkoxy (e.g., C1-6 alkoxy, such as perfluoroalkoxy); (4) C1-6 alkylsulfinyl; (5) C6-10 aryl; (6) amino; (7) C1-6 alk-C6-10 aryl; (8) azido; (9) C3-8 cycloalkyl; (10) C1-6 alk-C3-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C1-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1-20 thioalkoxy (e.g., C1-6 thioalkoxy); (17) —(CH2)qCO2RA′, where q is an integer from zero to four, and RA′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) hydrogen, and (d) C1-6 alk-C6-10 aryl; (18) —(CH2)qCONRB′RC′, where q is an integer from zero to four and where RB′ and RC′ are independently selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (19) —(CH2)qSO2RD′, where q is an integer from zero to four and where RD′ is selected from the group consisting of (a) alkyl, (b) C6-10 aryl, and (c) alk-C6-10 aryl; (20) —(CH2)qSO2NRE′RF′, where q is an integer from zero to four and where each of RE′ and RF′ is, independently, selected from the group consisting of (a) hydrogen, (b) C6-10 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (21) thiol; (22) C6-10 aryloxy; (23) C3-5 cycloalkoxy; (24) C6-10 aryl-C1-6 alkoxy; (25) C1-6 alk-C1-12 heterocyclyl (e.g., C1-6 alk-C1-12 heteroaryl); (26) C2-20 alkenyl; and (27) C2-20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a C1-alkaryl or a C1-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.

The term “arylalkoxy,” as used herein, represents an alkaryl group, as defined herein, attached to the parent molecular group through an oxygen atom. Exemplary unsubstituted alkoxyalkyl groups include from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C6-10 aryl-C1-6 alkoxy, C6-10 aryl-C1-10 alkoxy, or C6-10 aryl-C1-20 alkoxy). In some embodiments, the arylalkoxy group can be substituted with 1, 2, 3, or 4 substituents as defined herein

The term “aryloxy” represents a chemical substituent of formula —OR′, where R′ is an aryl group of 6 to 18 carbons, unless otherwise specified. In some embodiments, the aryl group can be substituted with 1, 2, 3, or 4 substituents as defined herein.

The term “aryloyl,” as used herein, represents an aryl group, as defined herein, that is attached to the parent molecular group through a carbonyl group. Exemplary unsubstituted aryloyl groups are of 7 to 11 carbons. In some embodiments, the aryl group can be substituted with 1, 2, 3, or 4 substituents as defined herein.

The term “azido” represents an —N3 group, which can also be represented as —N═N═N.

The term “bicyclic,” as used herein, refer to a structure having two rings, which may be aromatic or non-aromatic. Bicyclic structures include spirocyclyl groups, as defined herein, and two rings that share one or more bridges, where such bridges can include one atom or a chain including two, three, or more atoms. Exemplary bicyclic groups include a bicyclic carbocyclyl group, where the first and second rings are carbocyclyl groups, as defined herein; a bicyclic aryl groups, where the first and second rings are aryl groups, as defined herein; bicyclic heterocyclyl groups, where the first ring is a heterocyclyl group and the second ring is a carbocyclyl (e.g., aryl) or heterocyclyl (e.g., heteroaryl) group; and bicyclic heteroaryl groups, where the first ring is a heteroaryl group and the second ring is a carbocyclyl (e.g., aryl) or heterocyclyl (e.g., heteroaryl) group. In some embodiments, the bicyclic group can be substituted with 1, 2, 3, or 4 substituents as defined herein for cycloalkyl, heterocyclyl, and aryl groups.

The terms “carbocyclic” and “carbocyclyl,” as used herein, refer to an optionally substituted C3-12 monocyclic, bicyclic, or tricyclic structure in which the rings, which may be aromatic or non-aromatic, are formed by carbon atoms. Carbocyclic structures include cycloalkyl, cycloalkenyl, and aryl groups.

The term “carbamoyl,” as used herein, represents —C(O)—N(RN1)2, where the meaning of each RN1 is found in the definition of “amino” provided herein.

The term “carbamoylalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a carbamoyl group, as defined herein. The alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “carbamyl,” as used herein, refers to a carbamate group having the structure —NRN1C(═O)OR or —OC(═O)N(RN1)2, where the meaning of each RN1 is found in the definition of “amino” provided herein, and R is alkyl, cycloalkyl, alkcycloalkyl, aryl, alkaryl, heterocyclyl (e.g., heteroaryl), or alkheterocyclyl (e.g., alkheteroaryl), as defined herein.

The term “carbonyl,” as used herein, represents a C(O) group, which can also be represented as C═O.

The term “carboxyaldehyde” represents an acyl group having the structure —CHO.

The term “carboxy,” as used herein, means —CO2H.

The term “carboxyalkoxy,” as used herein, represents an alkoxy group, as defined herein, substituted by a carboxy group, as defined herein. The alkoxy group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for the alkyl group.

The term “carboxyalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a carboxy group, as defined herein. The alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “cyano,” as used herein, represents an —CN group.

The term “cycloalkoxy” represents a chemical substituent of formula —OR, where R is a C3-8 cycloalkyl group, as defined herein, unless otherwise specified. The cycloalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein. Exemplary unsubstituted cycloalkoxy groups are from 3 to 8 carbons. In some embodiment, the cycloalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “cycloalkyl,” as used herein represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.2.1]heptyl, and the like. When the cycloalkyl group includes one carbon-carbon double bond, the cycloalkyl group can be referred to as a “cycloalkenyl” group. Exemplary cycloalkenyl groups include cyclopentenyl, cyclohexenyl, and the like. The cycloalkyl groups of this invention can be optionally substituted with: (1) C1-7 acyl (e.g., carboxyaldehyde); (2) C1-20 alkyl (e.g., C1-6 alkyl, C1-6 alkoxy-C1-6 alkyl, C1-6 alkylsulfinyl-C1-6 alkyl, amino-C1-6 alkyl, azido-C1-6 alkyl, (carboxyaldehyde)-C1-6 alkyl, halo-C1-6 alkyl (e.g., perfluoroalkyl), hydroxy-C1-6 alkyl, nitro-C1-6 alkyl, or C1-6 thioalkoxy-C1-6 alkyl); (3) C1-20 alkoxy (e.g., C1-6 alkoxy, such as perfluoroalkoxy); (4) C1-6 alkylsulfinyl; (5) C6-10 aryl; (6) amino; (7) C1-6 alk-C6-10 aryl, (8) azido; (9) C3-8 cycloalkyl; (10) C1-6 alk-C3-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C1-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1-20 thioalkoxy (e.g., C1-6 thioalkoxy); (17) —(CH2)qCO2RA′, where q is an integer from zero to four, and RA′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) hydrogen, and (d) C1-6 alk-C6-10 aryl; (18) —(CH2)qCONRB′RC′, where q is an integer from zero to four and where RB′ and RC′ are independently selected from the group consisting of (a) hydrogen, (b) C6-10 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl, (19) —(CH2)qSO2RD′, where q is an integer from zero to four and where RD′ is selected from the group consisting of (a) C6-10 alkyl, (b) C6-10 aryl, and (c) C1-6 alk-C6-10 aryl; (20) —(CH2)qSO2NRE′RF′, where q is an integer from zero to four and where each of RE′ and RF′ is, independently, selected from the group consisting of (a) hydrogen, (b) C6-10 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (21) thiol; (22) C6-10 aryloxy; (23) C3-8 cycloalkoxy; (24) C6-10 aryl-C1-6 alkoxy; (25) C1-6 alk-C1-12 heterocyclyl (e.g., C1-6 alk-C1-12 heteroaryl); (26) oxo; (27) C2-20 alkenyl; and (28) C2-20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a C1-alkaryl or a C1-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.

The term “diastereomer,” as used herein means stereoisomers that are not mirror images of one another and are non-superimposable on one another.

The term “effective amount” of an agent, as used herein, is that amount sufficient to effect beneficial or desired results, for example, clinical results, and, as such, an “effective amount” depends upon the context in which it is being applied. For example, in the context of administering an agent that treats cancer, an effective amount of an agent is, for example, an amount sufficient to achieve treatment, as defined herein, of cancer, as compared to the response obtained without administration of the agent.

The term “enantiomer,” as used herein, means each individual optically active form of a compound of the invention, having an optical purity or enantiomeric excess (as determined by methods standard in the art) of at least 80% (i.e., at least 90% of one enantiomer and at most 10% of the other enantiomer), preferably at least 90% and more preferably at least 98%.

The term “halo,” as used herein, represents a halogen selected from bromine, chlorine, iodine, or fluorine.

The term “haloalkoxy,” as used herein, represents an alkoxy group, as defined herein, substituted by a halogen group (i.e., F, Cl, Br, or I). A haloalkoxy may be substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens. Haloalkoxy groups include perfluoroalkoxys (e.g., —OCF3), —OCHF2, —OCH2F, —OCCl3, —OCH2CH2Br, —OCH2CH(CH2CH2Br)CH3, and —OCHICH3. In some embodiments, the haloalkoxy group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups.

The term “haloalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a halogen group (i.e., F, Cl, Br, or I). A haloalkyl may be substituted with one, two, three, or, in the case of alkyl groups of two carbons or more, four halogens. Haloalkyl groups include perfluoroalkyls (e.g., —CF3), —CHF2, —CH2F, —CCl3, —CH2CH2Br, —CH2CH(CH2CH2Br)CH3, and —CHICH3. In some embodiments, the haloalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups.

The term “heteroalkylene,” as used herein, refers to an alkylene group, as defined herein, in which one or two of the constituent carbon atoms have each been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkylene group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkylene groups.

The term “heteroaryl,” as used herein, represents that subset of heterocyclyls, as defined herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the mono- or multicyclic ring system. Exemplary unsubstituted heteroaryl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. In some embodiment, the heteroaryl is substituted with 1, 2, 3, or 4 substituents groups as defined for a heterocyclyl group.

The term “heterocyclyl,” as used herein represents a 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. The 5-membered ring has zero to two double bonds, and the 6- and 7-membered rings have zero to three double bonds. Exemplary unsubstituted heterocyclyl groups are of 1 to 12 (e.g., 1 to 11, 1 to 10, 1 to 9, 2 to 12, 2 to 11, 2 to 10, or 2 to 9) carbons. The term “heterocyclyl” also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non-adjacent members of a monocyclic ring, e.g., a quinuclidinyl group. The term “heterocyclyl” includes bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three carbocyclic rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring, or another monocyclic heterocyclic ring, such as indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofiiryl, benzothienyl and the like. Examples of fused heterocyclyls include tropanes and 1,2,3,5,8,8a-hexahydroindolizine. Heterocyclics include pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, hom*opiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, indazolyl, quinolyl, isoquinolyl, quinoxalinyl, dihydroquinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzothiadiazolyl, furyl, thienyl, thiazolidinyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl (e.g., 1,2,3-oxadiazolyl), purinyl, thiadiazolyl (e.g., 1,2,3-thiadiazolyl), tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, dihydroindolyl, dihydroquinolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydroisoquinolyl, pyranyl, dihydropyranyl, dithiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, and the like, including dihydro and tetrahydro forms thereof, where one or more double bonds are reduced and replaced with hydrogens. Still other exemplary heterocyclyls include: 2,3,4,5-tetrahydro-2-oxo-oxazolyl; 2,3-dihydro-2-oxo-1H-imidazolyl; 2,3,4,5-tetrahydro-5-oxo-1H-pyrazolyl (e.g., 2,3,4,5-tetrahydro-2-phenyl-5-oxo-1H-pyrazolyl); 2,3,4,5-tetrahydro-2,4-dioxo-1H-imidazolyl (e.g., 2,3,4,5-tetrahydro-2,4-dioxo-5-methyl-5-phenyl-1H-imidazolyl); 2,3-dihydro-2-thioxo-1,3,4-oxadiazolyl (e.g., 2,3-dihydro-2-thioxo-5-phenyl-1,3,4-oxadiazolyl); 4,5-dihydro-5-oxo-1H-triazolyl (e.g., 4,5-dihydro-3-methyl-4-amino 5-oxo-1H-triazolyl); 1,2,3,4-tetrahydro-2,4-dioxopyridinyl (e.g., 1,2,3,4-tetrahydro-2,4-dioxo-3,3-diethylpyridinyl); 2,6-dioxo-piperidinyl (e.g., 2,6-dioxo-3-ethyl-3-phenylpiperidinyl); 1,6-dihydro-6-oxopyridiminyl; 1,6-dihydro-4-oxopyrimidinyl (e.g., 2-(methylthio)-1,6-dihydro-4-oxo-5-methylpyrimidin-1-yl); 1,2,3,4-tetrahydro-2,4-dioxopyrimidinyl (e.g., 1,2,3,4-tetrahydro-2,4-dioxo-3-ethylpyrimidinyl); 1,6-dihydro-6-oxo-pyridazinyl (e.g., 1,6-dihydro-6-oxo-3-ethylpyridazinyl); 1,6-dihydro-6-oxo-1,2,4-triazinyl (e.g., 1,6-dihydro-5-isopropyl-6-oxo-1,2,4-triazinyl); 2,3-dihydro-2-oxo-1H-indolyl (e.g., 3,3-dimethyl-2,3-dihydro-2-oxo-1H-indolyl and 2,3-dihydro-2-oxo-3,3′-spiropropane-1H-indol-1-yl); 1,3-dihydro-1-oxo-2H-iso-indolyl; 1,3-dihydro-1,3-dioxo-2H-iso-indolyl; 1H-benzopyrazolyl (e.g., 1-(ethoxycarbonyl)-1H-benzopyrazolyl); 2,3-dihydro-2-oxo-1H-benzimidazolyl (e.g., 3-ethyl-2,3-dihydro-2-oxo-1H-benzimidazolyl); 2,3-dihydro-2-oxo-benzoxazolyl (e.g., 5-chloro-2,3-dihydro-2-oxo-benzoxazolyl); 2,3-dihydro-2-oxo-benzoxazolyl; 2-oxo-2H-benzopyranyl; 1,4-benzodioxanyl; 1,3-benzodioxanyl; 2,3-dihydro-3-oxo,4H-1,3-benzothiazinyl; 3,4-dihydro-4-oxo-3H-quinazolinyl (e.g., 2-methyl-3,4-dihydro-4-oxo-3H-quinazolinyl); 1,2,3,4-tetrahydro-2,4-dioxo-3H-quinazolyl (e.g., 1-ethyl-1,2,3,4-tetrahydro-2,4-dioxo-3H-quinazolyl); 1,2,3,6-tetrahydro-2,6-dioxo-7H-purinyl (e.g., 1,2,3,6-tetrahydro-1,3-dimethyl-2,6-dioxo-7H-purinyl); 1,2,3,6-tetrahydro-2,6-dioxo-1H-purinyl (e.g., 1,2,3,6-tetrahydro-3,7-dimethyl-2,6-dioxo-1H-purinyl); 2-oxobenz[c,d]indolyl; 1,1-dioxo-2H-naphth[1,8-c,d]isothiazolyl; and 1,8-naphthylenedicarboxamido. Additional heterocyclics include 3,3a,4,5,6,6a-hexahydro-pyrrolo[3,4-b]pyrrol-(2H)-yl, and 2,5-diazabicyclo[2.2.1]heptan-2-yl, hom*opiperazinyl (or diazepanyl), tetrahydropyranyl, dithiazolyl, benzofuranyl, benzothienyl, oxepanyl, thiepanyl, azocanyl, oxecanyl, and thiocanyl. Heterocyclic groups also include groups of the formula

where

E′ is selected from the group consisting of —N— and —CH—; F′ is selected from the group consisting of —N═CH—, —NH—CH2—, —NH—C(O)—, —NH—, —CH═N—, —CH2—NH—, —C(O)—NH—, —CH═CH—, —CH2—, —CH2CH2—, —CH2O—, —OCH2—, —O—, and —S—; and G′ is selected from the group consisting of —CH— and —N—. Any of the heterocyclyl groups mentioned herein may be optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of: (1) C1-7 acyl (e.g., carboxyaldehyde); (2) C1-20 alkyl (e.g., C1-6 alkyl, C1-6 alkoxy-C1-6 alkyl, C1-6 alkylsulfinyl-C1-6 alkyl, amino-C1-6 alkyl, azido-C1-6 alkyl, (carboxyaldehyde)-C1-6 alkyl, halo-C1-6 alkyl (e.g., perfluoroalkyl), hydroxy-C1-6 alkyl, nitro-C1-6 alkyl, or C1-6 thioalkoxy-C1-6 alkyl); (3) C1-20 alkoxy (e.g., C1-6 alkoxy, such as perfluoroalkoxy); (4) C1-6 alkylsulfinyl; (5) C6-10 aryl; (6) amino; (7) C1-6 alk-C6-10 aryl; (8) azido; (9) C3-8 cycloalkyl; (10) C1-6 alk-C3-8 cycloalkyl; (11) halo; (12) C1-12 heterocyclyl (e.g., C2-12 heteroaryl); (13) (C1-12 heterocyclyl)oxy; (14) hydroxy; (15) nitro; (16) C1-20 thioalkoxy (e.g., C1-6 thioalkoxy); (17) —(CH2)qCO2RA′, where q is an integer from zero to four, and RA′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, (c) hydrogen, and (d) C1-6 alk-C6-10 aryl; (18) —(CH2)qCONRB′RC′, where q is an integer from zero to four and where RB′ and RC′ are independently selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (19) —(CH2)qSO2RD′, where q is an integer from zero to four and where RD′ is selected from the group consisting of (a) C1-6 alkyl, (b) C6-10 aryl, and (c) C1-6 alk-C6-10 aryl; (20) —(CH2)qSO2NRE′RF′, where q is an integer from zero to four and where each of RE′ and RF′ is, independently, selected from the group consisting of (a) hydrogen, (b) C1-6 alkyl, (c) C6-10 aryl, and (d) C1-6 alk-C6-10 aryl; (21) thiol; (22) C6-10 aryloxy; (23) C3-8 cycloalkoxy; (24) arylalkoxy; (25) C1-6 alk-C1-12 heterocyclyl (e.g., C1-6 alk-C1-12 heteroaryl); (26) oxo; (27) (C1-12 heterocyclyl)imino; (28) C2-20 alkenyl; and (29) C2-20 alkynyl. In some embodiments, each of these groups can be further substituted as described herein. For example, the alkylene group of a C1-alkaryl or a C1-alkheterocyclyl can be further substituted with an oxo group to afford the respective aryloyl and (heterocyclyl)oyl substituent group.

The term “(heterocyclyl)imino,” as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an imino group. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as defined herein.

The term “(heterocyclyl)oxy,” as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through an oxygen atom. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as defined herein.

The term “(heterocyclyl)oyl,” as used herein, represents a heterocyclyl group, as defined herein, attached to the parent molecular group through a carbonyl group. In some embodiments, the heterocyclyl group can be substituted with 1, 2, 3, or 4 substituent groups as defined herein.

The term “hydrocarbon,” as used herein, represents a group consisting only of carbon and hydrogen atoms.

The term “hydroxy,” as used herein, represents an —OH group.

The term “hydroxyalkenyl,” as used herein, represents an alkenyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group, and is exemplified by dihydroxypropenyl, hydroxyisopentenyl, and the like.

The term “hydroxyalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group, and is exemplified by hydroxymethyl, dihydroxypropyl, and the like.

The term “isomer,” as used herein, means any tautomer, stereoisomer, enantiomer, or diastereomer of any compound of the invention. It is recognized that the compounds of the invention can have one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric E/Z isomers) or diastereomers (e.g., enantiomers (i.e., (+) or (−)) or cis/trans isomers). According to the invention, the chemical structures depicted herein, and therefore the compounds of the invention, encompass all of the corresponding stereoisomers, that is, both the stereomerically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and enantiomeric and stereoisomeric mixtures, e.g., racemates. Enantiomeric and stereoisomeric mixtures of compounds of the invention can typically be resolved into their component enantiomers or stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Enantiomers and stereoisomers can also be obtained from stereomerically or enantiomerically pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.

The term “N-protected amino,” as used herein, refers to an amino group, as defined herein, to which is attached one or two N-protecting groups, as defined herein.

The term “N-protecting group,” as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis,” 3rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference. N-protecting groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, α-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine, and the like; sulfonyl-containing groups such as benzenesulfonyl, p-toluenesulfonyl, and the like; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyloxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)-1-methylethoxycarbonyl, α,α-dimethyl-3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, phenylthiocarbonyl, and the like, alkaryl groups such as benzyl, triphenylmethyl, benzyloxymethyl, and the like and say (groups, such as trimethylsilyl, and the like. Preferred N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).

The term “nitro,” as used herein, represents an —NO2 group.

The term “oxo” as used herein, represents ═O.

The term “perfluoroalkyl,” as used herein, represents an alkyl group, as defined herein, where each hydrogen radical bound to the alkyl group has been replaced by a fluoride radical. Perfluoroalkyl groups are exemplified by trifluoromethyl, pentafluoroethyl, and the like.

The term “perfluoroalkoxy,” as used herein, represents an alkoxy group, as defined herein, where each hydrogen radical bound to the alkoxy group has been replaced by a fluoride radical. Perfluoroalkoxy groups are exemplified by trifluoromethoxy, pentafluoroethoxy, and the like.

The term “spirocyclyl,” as used herein, represents a C2-7 alkylene diradical, both ends of which are bonded to the same carbon atom of the parent group to form a spirocyclic group, and also a C1-6 heteroalkylene diradical, both ends of which are bonded to the same atom. The heteroalkylene radical forming the spirocyclyl group can containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. In some embodiments, the spirocyclyl group includes one to seven carbons, excluding the carbon atom to which the diradical is attached. The spirocyclyl groups of the invention may be optionally substituted with 1, 2, 3, or 4 substituents provided herein as optional substituents for cycloalkyl and/or heterocyclyl groups.

The term “stereoisomer,” as used herein, refers to all possible different isomeric as well as conformational forms which a compound may possess (e.g., a compound of any formula described herein), in particular all possible stereochemically and conformationally isomeric forms, all diastereomers, enantiomers and/or conformers of the basic molecular structure. Some compounds of the present invention may exist in different tautomeric forms, all of the latter being included within the scope of the present invention.

The term “sulfoalkyl,” as used herein, represents an alkyl group, as defined herein, substituted by a sulfo group of —SO3H. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “sulfonyl,” as used herein, represents an —S(O)2— group.

The term “thioalkaryl,” as used herein, represents a chemical substituent of formula —SR, where R is an alkaryl group. In some embodiments, the alkaryl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “thioalkheterocyclyl,” as used herein, represents a chemical substituent of formula —SR, where R is an alkheterocyclyl group. In some embodiments, the alkheterocyclyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “thioalkoxy,” as used herein, represents a chemical substituent of formula —SR, where R is an alkyl group, as defined herein. In some embodiments, the alkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein.

The term “thiol” represents an —SH group.

Compound: As used herein, the term “compound,” is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted.

The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C═N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present disclosure. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms.

Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples prototropic tautomers include ketone-enol pairs, amide-imidic acid pairs, lactam-lactim pairs, amide-imidic acid pairs, enamine-imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H-1,2,4-triazole, 1H- and 2H-isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.

Compounds of the present disclosure also include all of the isotopes of the atoms occurring in the intermediate or final compounds. “Isotopes” refers to atoms having the same atomic number but different mass numbers resulting from a different number of neutrons in the nuclei. For example, isotopes of hydrogen include tritium and deuterium.

The compounds and salts of the present disclosure can be prepared in combination with solvent or water molecules to form solvates and hydrates by routine methods.

Condition: As used herein, the term “condition” refers to a disorder that presents with observable symptoms.

Conserved: As used herein, the term “conserved” refers to nucleotides or amino acid residues of a polynucleotide sequence or polypeptide sequence, respectively, that are those that occur unaltered in the same position of two or more sequences being compared. Nucleotides or amino acids that are relatively conserved are those that are conserved amongst more related sequences than nucleotides or amino acids appearing elsewhere in the sequences.

In some embodiments, two or more sequences are said to be “completely conserved” if they are 100% identical to one another. In some embodiments, two or more sequences are said to be “highly conserved” if they are at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be “highly conserved” if they are about 70% identical, about 80% identical, about 90% identical, about 95%, about 98%, or about 99% identical to one another. In some embodiments, two or more sequences are said to be “conserved” if they are at least 30% identical, at least 40% identical, at least 50% identical, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be “conserved” if they are about 30% identical, about 40% identical, about 50% identical, about 60% identical, about 70% identical, about 80% identical, about 90% identical, about 95% identical, about 98% identical, or about 99% identical to one another. Conservation of sequence may apply to the entire length of an oligonucleotide or polypeptide or may apply to a portion, region or feature thereof.

Cyclic or Cyclized: As used herein, the term “cyclic” refers to the presence of a continuous loop. Cyclic molecules need not be circular, only joined to form an unbroken chain of subunits. Cyclic molecules such as the engineered RNA or mRNA of the present invention may be single units or multimers or comprise one or more components of a complex or higher order structure.

Cytostatic: As used herein, “cytostatic” refers to inhibiting, reducing, suppressing the growth, division, or multiplication of a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.

Cytotoxic: As used herein, “cytotoxic” refers to killing or causing injurious, toxic, or deadly effect on a cell (e.g., a mammalian cell (e.g., a human cell)), bacterium, virus, fungus, protozoan, parasite, prion, or a combination thereof.

Delivery: As used herein, “delivery” refers to the act or manner of delivering a compound, substance, entity, moiety, cargo or payload.

Delivery Agent: As used herein, “delivery agent” refers to any substance which facilitates, at least in part, the in vivo delivery of signal-sensor polynucleotide, primary construct or mmRNA to targeted cells.

Destabilized: As used herein, the term “destable,” “destabilize,” or “destabilizing region” means a region or molecule that is less stable than a starting, wild-type or native form of the same region or molecule.

Detectable label: As used herein, “detectable label” refers to one or more markers, signals, or moieties which are attached, incorporated or associated with another entity that is readily detected by methods known in the art including radiography, fluorescence, chemiluminescence, enzymatic activity, absorbance and the like. Detectable labels include radioisotopes, fluorophores, chromophores, enzymes, dyes, metal ions, ligands such as biotin, avidin, streptavidin and haptens, quantum dots, and the like. Detectable labels may be located at any position in the peptides or proteins disclosed herein. They may be within the amino acids, the peptides, or proteins, or located at the N- or C-termini.

Disease: As used herein, the term “disease” refers to an abnormal condition affecting the body of an organism often showing specific bodily symptoms.

Disorder: As used herein, the term “disorder,” refers to a disruption of or an interference with normal functions or established systems of the body.

Digest: As used herein, the term “digest” means to break apart into smaller pieces or components. When referring to polypeptides or proteins, digestion results in the production of peptides.

Distal: As used herein, the term “distal” means situated away from the center or away from a point or region of interest.

Dose splitting factor (DSF)-ratio of PUD of dose split treatment divided by PUD of total daily dose or single unit dose. The value is derived from comparison of dosing regimens groups.

Encoded protein cleavage signal: As used herein, “encoded protein cleavage signal” refers to the nucleotide sequence which encodes a protein cleavage signal.

Engineered: As used herein, embodiments of the invention are “engineered” when they are designed to have a feature or property, whether structural or chemical, that varies from a starting point, wild type or native molecule.

Exosome: As used herein, “exosome” is a vesicle secreted by mammalian cells or a complex involved in RNA degradation.

Expression: As used herein, “expression” of a nucleic acid sequence refers to one or more of the following events: (I) production of an RNA template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5′ cap formation, and/or 3′ end processing); (3) translation of an RNA into a polypeptide or protein; and (4) post-translational modification of a polypeptide or protein.

Feature: As used herein, a “feature” refers to a characteristic, a property, or a distinctive element.

Formulation: As used herein, a “formulation” includes at least a signal-sensor polynucleotide, primary construct or mmRNA and a delivery agent.

Fragment: A “fragment,” as used herein, refers to a portion. For example, fragments of proteins may comprise polypeptides obtained by digesting full-length protein isolated from cultured cells.

Functional: As used herein, a “functional” biological molecule is a biological molecule in a form in which it exhibits a property and/or activity by which it is characterized.

Genotype: As used herein, “genotype” refers to the change in the genotype, or genetic makeup, of a subject, cell, tissue, organ and/or organism.

hom*ology: As used herein, the term “hom*ology” refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. In some embodiments, polymeric molecules are considered to be “hom*ologous” to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or similar. The term “hom*ologous” necessarily refers to a comparison between at least two sequences (polynucleotide or polypeptide sequences). In accordance with the invention, two polynucleotide sequences are considered to be hom*ologous if the polypeptides they encode are at least about 50%, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino acids. In some embodiments, hom*ologous polynucleotide sequences are characterized by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. For polynucleotide sequences less than 60 nucleotides in length, hom*ology is determined by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. In accordance with the invention, two protein sequences are considered to be hom*ologous if the proteins are at least about 50%, 60%, 70%, 80%, or 90% identical for at least one stretch of at least about 20 amino acids.

Identity: As used herein, the term “identity” refers to the overall relatedness between polymeric molecules, e.g., between oligonucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes). In certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the length of the reference sequence. The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; each of which is incorporated herein by reference. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS, 1989, 4:11-17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix.

Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H., and Lipman, D., SIAM J Applied Math., 48:1073 (1988); incorporated herein by reference. Techniques for determining identity are codified in publicly available computer programs. Exemplary computer software to determine hom*ology between two sequences include, but are not limited to, GCG program package, Devereux, J., et al., Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altschul, S. F. et al., J. Molec. Biol., 215, 403 (1990)).

Inhibit expression of a gene: As used herein, the phrase “inhibit expression of a gene” means to cause a reduction in the amount of an expression product of the gene. The expression product can be an RNA transcribed from the gene (e.g., an mRNA) or a polypeptide translated from an mRNA transcribed from the gene. Typically a reduction in the level of an mRNA results in a reduction in the level of a polypeptide translated therefrom. The level of expression may be determined using standard techniques for measuring mRNA or protein.

In vitro: As used herein, the term “in vitro” refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).

In vivo: As used herein, the term “in vivo” refers to events that occur within an organism (e.g., animal, plant, or microbe or cell or tissue thereof).

Isolated: As used herein, the term “isolated” refers to a substance or entity that has been separated from at least some of the components with which it was associated (whether in nature or in an experimental setting). Isolated substances may have varying levels of purity in reference to the substances from which they have been associated. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some embodiments, isolated agents are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is “pure” if it is substantially free of other components. Substantially isolated: By “substantially isolated” is meant that the compound is substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compound of the present disclosure. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art.

Linker: As used herein, a linker refers to a group of atoms, e.g., 10-1,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, alkylamino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine. The linker can be attached to a modified nucleoside or nucleotide on the nucleobase or sugar moiety at a first end, and to a payload, e.g., a detectable or therapeutic agent, at a second end. The linker may be of sufficient length as to not interfere with incorporation into a nucleic acid sequence. The linker can be used for any useful purpose, such as to form mmRNA multimers (e.g., through linkage of two or more signal-sensor polynucleotides, primary constructs, or mmRNA molecules) or mmRNA conjugates, as well as to administer a payload, as described herein. Examples of chemical groups that can be incorporated into the linker include, but are not limited to, alkyl, alkenyl, alkynyl, amido, amino, ether, thioether, ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be optionally substituted, as described herein. Examples of linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols (e.g., ethylene or propylene glycol monomeric units, e.g., diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, or tetraethylene glycol), and dextran polymers, Other examples include, but are not limited to, cleavable moieties within the linker, such as, for example, a disulfide bond (—S—S—) or an azo bond (—N═N—), which can be cleaved using a reducing agent or photolysis. Non-limiting examples of a selectively cleavable bond include an amido bond can be cleaved for example by the use of tris(2-carboxyethyl)phosphine (TCEP), or other reducing agents, and/or photolysis, as well as an ester bond can be cleaved for example by acidic or basic hydrolysis.

Metastasis: As used herein, the term “metastasis” means the process by which cancer spreads from the place at which it first arose as a primary tumor to distant locations in the body.

Method of Treating: The phrase “a method of treating” or its equivalent, when applied to, for example, cancer refers to a procedure or course of action that is designed to reduce or eliminate the number of cancer cells, prevent the increase in the number of cancer cells, or to alleviate the symptoms of a cancer in a subject. A method of treating cancer or another oncology-related disorder does not necessarily mean that the cancer cells or other disorder will, in fact, be completely eliminated, that the number of cells or disorder will, in fact, be reduced, or that the symptoms of a cancer or other disorder will, in fact, be alleviated. Often, a method of treating cancer will be performed even with a low likelihood of success, but which, given the medical history and estimated survival expectancy of a subject, is nevertheless deemed an overall beneficial course of action.

MicroRNA (miRNA) binding site: As used herein, a microRNA (miRNA) binding site represents a nucleotide location or region of a nucleic acid transcript to which at least the “seed” region of a miRNA binds.

Modified: As used herein “modified” refers to a changed state or structure of a molecule of the invention. Molecules may be modified in many ways including chemically, structurally, and functionally. In one embodiment, the mRNA molecules of the present invention are modified by the introduction of non-natural nucleosides and/or nucleotides, e.g., as it relates to the natural ribonucleotides A, U, G, and C. Noncanonical nucleotides such as the cap structures are not considered “modified” although they differ from the chemical structure of the A, C, G, U ribonucleotides.

Mucus: As used herein, “mucus” refers to the natural substance that is viscous and comprises mucin glycoproteins.

Naturally occurring: As used herein, “naturally occurring” means existing in nature without artificial aid.

Non-human vertebrate: As used herein, a “non human vertebrate” includes all vertebrates except hom*o sapiens, including wild and domesticated species. Examples of non-human vertebrates include, but are not limited to, mammals, such as alpaca, banteng, bison, camel, cat, cattle, deer, dog, donkey, gayal, goat, guinea pig, horse, llama, mule, pig, rabbit, reindeer, sheep water buffalo, and yak.

Off-target: As used herein, “off target” refers to any unintended effect on any one or more target, gene, or cellular transcript.

Oncology-related: As used herein, the term “oncology-related” refers to any disease, disorder, condition, treatment, process, substance or compound related to any aspect of one or more hyperproliferative diseases, disorders and/or conditions including, but not limited to, cancer.

Open reading frame: As used herein, “open reading frame” or “ORF” refers to a sequence which does not contain a stop codon in a given reading frame.

Operably linked: As used herein, the phrase “operably linked” refers to a functional connection between two or more molecules, constructs, transcripts, entities, moieties or the like.

Paratope: As used herein, a “paratope” refers to the antigen-binding site of an antibody.

Patient: As used herein, “patient” refers to a subject who may seek or be in need of treatment, requires treatment, is receiving treatment, will receive treatment, or a subject who is under care by a trained professional for a particular disease or condition.

Optionally substituted: Herein a phrase of the form “optionally substituted X” (e.g., optionally substituted alkyl) is intended to be equivalent to “X, wherein X is optionally substituted” (e.g., “alkyl, wherein said alkyl is optionally substituted”). It is not intended to mean that the feature “X” (e.g. alkyl) per se is optional.

Peptide: As used herein, “peptide” is less than or equal to 50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.

Pharmaceutical composition: The phrase “pharmaceutical composition” refers to a composition that alters the etiology of a disease, disorder and/or condition.

Pharmaceutically acceptable: The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

Pharmaceutically acceptable excipients: The phrase “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

Pharmaceutically acceptable salts: The present disclosure also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, “pharmaceutically acceptable salts” refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form (e.g., by reacting the free base group with a suitable organic acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. The pharmaceutically acceptable salts of the present disclosure include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present disclosure can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P. H. Stahl and C. G. Wermuth (eds.), Wiley-VCH, 2008, and Berge et al., Journal of Pharmaceutical Science, 66, 1-19 (1977), each of which is incorporated herein by reference in its entirety.

Pharmaceutically acceptable solvate: The term “pharmaceutically acceptable solvate,” as used herein, means a compound of the invention wherein molecules of a suitable solvent are incorporated in the crystal lattice. A suitable solvent is physiologically tolerable at the dosage administered. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-, and tri-hydrates), N-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), N,N′-dimethylformamide (DMF), N,N′-dimethylacetamide (DMAC), 1,3-dimethyl-2-imidazolidinone (DMEU), 1,3-dimethyl-3,4,5,6-tetrahydro-2-(1H)-pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the solvate is referred to as a “hydrate.”

Pharmaco*kinetic: As used herein, “pharmaco*kinetic” refers to any one or more properties of a molecule or compound as it relates to the determination of the fate of substances administered to a living organism. Pharmaco*kinetics is divided into several areas including the extent and rate of absorption, distribution, metabolism and excretion. This is commonly referred to as ADME where: (A) Absorption is the process of a substance entering the blood circulation; (D) Distribution is the dispersion or dissemination of substances throughout the fluids and tissues of the body; (M) Metabolism (or Biotransformation) is the irreversible transformation of parent compounds into daughter metabolites; and (E) Excretion (or Elimination) refers to the elimination of the substances from the body. In rare cases, some drugs irreversibly accumulate in body tissue.

Phenotype: As used herein, “phenotype” refers to the set of observable characteristics of a subject, cell, tissue, organ and/or organism.

Physicochemical: As used herein, “physicochemical” means of or relating to a physical and/or chemical property.

Preventing: As used herein, the term “preventing” refers to partially or completely delaying onset of an infection, disease, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying progression from an infection, a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the infection, the disease, disorder, and/or condition.

Prodrug: The present disclosure also includes prodrugs of the compounds described herein. As used herein, “prodrugs” refer to any substance, molecule or entity which is in a form predicate for that substance, molecule or entity to act as a therapeutic upon chemical or physical alteration. Prodrugs may by covalently bonded or sequestered in some way and which release or are converted into the active drug moiety prior to, upon or after administered to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.

Proliferate: As used herein, the term “proliferate” means to grow, expand or increase or cause to grow, expand or increase rapidly. “Proliferative” means having the ability to proliferate. “Anti-proliferative” means having properties counter to or inapposite to proliferative properties.

Protein cleavage site: As used herein, “protein cleavage site” refers to a site where controlled cleavage of the amino acid chain can be accomplished by chemical, enzymatic or photochemical means.

Protein cleavage signal: As used herein “protein cleavage signal” refers to at least one amino acid that flags or marks a polypeptide for cleavage.

Progression: As used herein, the term “progression” (e.g., cancer progression) means the advancement or worsening of or toward a disease or condition.

Protein of interest: As used herein, the terms “proteins of interest” or “desired proteins” include those provided herein and fragments, mutants, variants, and alterations thereof.

Proximal: As used herein, the term “proximal” means situated nearer to the center or to a point or region of interest.

Pseudouridine: As used herein, pseudouridine refers to the C-glycoside isomer of the nucleoside uridine. A “pseudouridine analog” is any modification, variant, isoform or derivative of pseudouridine. For example, pseudouridine analogs include but are not limited to 1-carboxymethyl-pseudouridine, 1-propynyl-pseudouridine, 1-taurinomethyl-pseudouridine, 1-taurinomethyl-4-thio-pseudouridine, 1-methyl-pseudouridine (m1ψ), 1-methyl-4-thio-pseudouridine (m1s4ψ), 4-thio-1-methyl-pseudouridine, 3-methyl-pseudouridine (m3ψ), 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydropseudouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, N1-methyl-pseudouridine, 1-methyl-3-(3-amino-3-carboxypropyl)pseudouridine (acp3 ψ), and 2′-O-methyl-pseudouridine (ψm).

Purified: As used herein, “purify,” “purified,” “purification” means to make substantially pure or clear from unwanted components, material defilement, admixture or imperfection.

Regression: As used herein, the term “regression” or “degree of regression” refers to the reversal, either phenotypically or genotypically, of a cancer progression. Slowing or stopping cancer progression may be considered regression.

Reducing the effect: As used herein, the phrase “reducing the effect” when referring to symptoms, means reducing, eliminating or alleviating the symptom in the subject. It does not necessarily mean that the symptom will, in fact, be completely eliminated, reduced or alleviated.

Sample: As used herein, the term “sample” or “biological sample” refers to a subset of its tissues, cells or component parts (e.g. body fluids, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vagin*l fluid and sem*n). A sample further may include a hom*ogenate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs. A sample further refers to a medium, such as a nutrient broth or gel, which may contain cellular components, such as proteins or nucleic acid molecule.

Side effect: As used herein, the phrase “side effect” refers to a secondary effect of treatment.

Signal Peptide Sequences: As used herein, the phrase “signal peptide sequences” refers to a sequence which can direct the transport or localization of a protein.

Signal-sensor polynucleotide: As used herein, “signal-sensor polynucleotides” are nucleic acid transcripts which encode one or more oncology-related polypeptides of interest that, when translated, delivers a “signal” to the cell (cancer or noncancerous) which results in the therapeutic benefit to the organism of either being detrimental to the cancer cell or beneficial to normal cells or both detrimental to cancer cells and advantageous to normal cells. The signal-sensor polynucleotides may optionally further comprise a sequence (translatable or not) which “senses” the microenvironment of the polynucleotide and alters (a) the function or phenotypic outcome associated with the peptide or protein which is translated, (b) the expression level of the signal-sensor polynucleotide, and/or both.

Single unit dose: As used herein, a “single unit dose” is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single administration event.

Similarity: As used herein, the term “similarity” refers to the overall relatedness between polymeric molecules, e.g. between polynucleotide molecules (e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art.

Skin: The term “skin” is the thin layer of tissue forming the natural outer covering of the body of a subject and includes the epidermis and the dermis. The dermis is the thick layer of living tissue below the epidermis which is the surface epithelium of the skin.

Split dose: As used herein, a “split dose” is the division of single unit dose or total daily dose into two or more doses.

Stable: As used herein “stable” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.

Stabilized: As used herein, the term “stabilize”, “stabilized,” “stabilized region” means to make or become stable.

Subject: As used herein, the term “subject” or “patient” refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.

Substantially: As used herein, the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term “substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.

Substantially equal: As used herein as it relates to time differences between doses, the term means plus/minus 2%.

Substantially simultaneously: As used herein and as it relates to plurality of doses, the term means within 2 seconds.

Suffering from: An individual who is “suffering from” a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.

Susceptible to: An individual who is “susceptible to” a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition but harbors a propensity to develop a disease or its symptoms. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition (for example, cancer) may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.

Symptom: As used herein, the term “symptom” is a signal of a disease, disorder and/or condition. For example, symptoms may be felt or noticed by the subject who has them but may not be easily accessed by looking at a subject's outward appearance or behaviors. Examples of symptoms include, but are not limited to, weakness, aches and pains, fever, fatigue, weight loss, blood clots, increased blood calcium levels, low white blood cell count, short of breath, dizziness, headaches, hyperpigmentation, jaundice, erthema, pruritis, excessive hair growth, change in bowel habits, change in bladder function, long-lasting sores, white patches inside the mouth, white spots on the tongue, unusual bleeding or discharge, thickening or lump on parts of the body, indigestion, trouble swallowing, changes in warts or moles, change in new skin and nagging cough or hoarseness.

Synthetic: The term “synthetic” means produced, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present invention may be chemical or enzymatic.

Targeted Cells: As used herein, “targeted cells” refers to any one or more cells of interest. The cells may be found in vitro, in vivo, in situ or in the tissue or organ of an organism. The organism may be an animal, preferably a mammal, more preferably a human and most preferably a patient.

Therapeutic Agent: The term “therapeutic agent” refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.

Therapeutically effective amount: As used herein, the term “therapeutically effective amount” means an amount of an agent to be delivered (e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is sufficient, when administered to a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.

Therapeutically effective outcome: As used herein, the term “therapeutically effective outcome” means an outcome that is sufficient in a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.

Total daily dose: As used herein, a “total daily dose” is an amount given or prescribed in 24 hr period. It may be administered as a single unit dose.

Transcription factor: As used herein, the term “transcription factor” refers to a DNA-binding protein that regulates transcription of DNA into RNA, for example, by activation or repression of transcription. Some transcription factors effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcription factors may regulate transcription of a target gene alone or in a complex with other molecules.

Treating: As used herein, the term “treating” refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular infection, disease, disorder, and/or condition. For example, “treating” cancer may refer to inhibiting survival, growth, and/or spread of a tumor. Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.

Tumor: As used herein, a “tumor” is an abnormal growth of tissue, whether benign or malignant.

Tumor growth: As used herein, the term “tumor growth” or “tumor metastases” means an increased mass or volume of the tumor or expansion of the tumor distribution.

Unmodified: As used herein, “unmodified” refers to any substance, compound or molecule prior to being changed in any way. Unmodified may, but does not always, refer to the wild type or native form of a biomolecule. Molecules may undergo a series of modifications whereby each modified molecule may serve as the “unmodified” starting molecule for a subsequent modification.

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments in accordance with the invention described herein. The scope of the present invention is not intended to be limited to the above Description, but rather is as set forth in the appended claims.

In the claims, articles such as “a,” “an,” and “the” may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.

It is also noted that the term “comprising” is intended to be open and permits but does not require the inclusion of additional elements or steps. When the term “comprising” is used herein, the term “consisting of” is thus also encompassed and disclosed.

Where ranges are given, endpoints are included. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.

In addition, it is to be understood that any particular embodiment of the present invention that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular embodiment of the compositions of the invention (e.g., any nucleic acid or protein encoded thereby; any method of production; any method of use; etc.) can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art.

All cited sources, for example, references, publications, databases, database entries, and art cited herein, are incorporated into this application by reference, even if not expressly stated in the citation. In case of conflicting statements of a cited source and the instant application, the statement in the instant application shall control.

Section and table headings are not intended to be limiting.

Modified signal-sensor mRNAs (mmRNA) according to the invention may be made using standard laboratory methods and materials. The open reading frame (ORF) of the gene of interest may be flanked by a 5′ untranslated region (UTR) which may contain a strong Kozak translational initiation signal and/or an alpha-globin 3′ UTR which may include an oligo(dT) sequence for templated addition of a poly-A tail. The modified mRNAs may be modified to reduce the cellular innate immune response. The modifications to reduce the cellular response may include pseudouridine (ψ) and 5-methyl-cytidine (5meC, 5mc or m5C). (See, Kariko K et al. Immunity 23:165-75 (2005), Kariko K et al. Mol Ther 16:1833-40 (2008), Anderson B R et al. NAR (2010); herein incorporated by reference).

The ORF may also include various upstream or downstream additions (such as, but not limited to, β-globin, tags, etc.) may be ordered from an optimization service such as, but limited to, DNA2.0 (Menlo Park, Calif.) and may contain multiple cloning sites which may have XbaI recognition. Upon receipt of the construct, it may be reconstituted and transformed into chemically competent E. coli.

For the present invention, NEB DH5-alpha Competent E. coli may be used. Transformations are performed according to NEB instructions using 100 ng of plasmid. The protocol is as follows:

Thaw a tube of NEB 5-alpha Competent E. coli cells on ice for 10 minutes.

Add 1-5 μl containing 1 pg-100 ng of plasmid DNA to the cell mixture. Carefully flick the tube 4-5 times to mix cells and DNA. Do not vortex.

Place the mixture on ice for 30 minutes. Do not mix.

Heat shock at 42° C. for exactly 30 seconds. Do not mix.

Place on ice for 5 minutes. Do not mix.

Pipette 950 μl of room temperature SOC into the mixture.

Place at 37° C. for 60 minutes. Shake vigorously (250 rpm) or rotate.

Warm selection plates to 37° C.

Mix the cells thoroughly by flicking the tube and inverting.

Spread 50-100 μl of each dilution onto a selection plate and incubate overnight at 37° C. Alternatively, incubate at 30° C. for 24-36 hours or 25° C. for 48 hours.

A single colony is then used to inoculate 5 ml of LB growth media using the appropriate antibiotic and then allowed to grow (250 RPM, 37° C.) for 5 hours. This is then used to inoculate a 200 ml culture medium and allowed to grow overnight under the same conditions.

To isolate the plasmid (up to 850 μg), a maxi prep is performed using the Invitrogen PURELINK™ HiPure Maxiprep Kit (Carlsbad, Calif.), following the manufacturer's instructions.

In order to generate cDNA for In Vitro Transcription (IVT), the plasmid is first linearized using a restriction enzyme such as XbaI. A typical restriction digest with XbaI will comprise the following: Plasmid 1.0 μg; 10× Buffer 1.0 μl; XbaI 1.5 μl; dH2O up to 10 μl; incubated at 37° C. for 1 hr. If performing at lab scale (<5 μg), the reaction is cleaned up using Invitrogen's PURELINK™ PCR Micro Kit (Carlsbad, Calif.) per manufacturer's instructions. Larger scale purifications may need to be done with a product that has a larger load capacity such as Invitrogen's standard PURELINK™ PCR Kit (Carlsbad, Calif.). Following the cleanup, the linearized vector is quantified using the NanoDrop and analyzed to confirm linearization using agarose gel electrophoresis.

As a non-limiting example, G-CSF may represent the polypeptide of interest. Sequences used in the steps outlined in Examples 1-5 are shown in Table 12. It should be noted that the start codon (ATG) has been underlined in each sequence of Table 12.

TABLE 12
G-CSF Sequences
SEQ ID NODescription
6592cDNAsequence:
ATGGCTGGACCTGCCACCCAGAGCCCCATGAAGCTGATGGCCCTGCAGCT
GCTGCTGTGGCACAGTGCACTCTGGACAGTGCAGGAAGCCACCCCCCTGG
GCCCTGCCAGCTCCCTGCCCCAGAGCTTCCTGCTCAAGTGCTTAGAGCAA
GTGAGGAAGATCCAGGGCGATGGCGCAGCGCTCCAGGAGAAGCTGTGTG
CCACCTACAAGCTGTGCCACCCCGAGGAGCTGGTGCTGCTCGGACACTCT
CTGGGCATCCCCTGGGCTCCCCTGAGCAGCTGCCCCAGCCAGGCCCTGCA
GCTGGCAGGCTGCTTGAGCCAACTCCATAGCGGCCTTTTCCTCTACCAGG
GGCTCCTGCAGGCCCTGGAAGGGATCTCCCCCGAGTTGGGTCCCACCTTG
GACACACTGCAGCTGGACGTCGCCGACTTTGCCACCACCATCTGGCAGCA
GATGGAAGAACTGGGAATGGCCCCTGCCCTGCAGCCCACCCAGGGTGCC
ATGCCGGCCTTCGCCTCTGCTTTCCAGCGCCGGGCAGGAGGGGTCCTGGT
TGCCTCCCATCTGCAGAGCTTCCTGGAGGTGTCGTACCGCGTTCTACGCC
ACCTTGCCCAGCCCTGA
6593cDNA having T7 polymerase site, AfeI and Xba
restriction site:
TAATACGACTCACTATA
GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACC
ATGGCTGGACCTGCCACCCAGAGCCCCATGAAGCTGATGGCCCTGCAGCT
GCTGCTGTGGCACAGTGCACTCTGGACAGTGCAGGAAGCCACCCCCCTGG
GCCCTGCCAGCTCCCTGCCCCAGAGCTTCCTGCTCAAGTGCTTAGAGCAA
GTGAGGAAGATCCAGGGCGATGGCGCAGCGCTCCAGGAGAAGCTGTGTG
CCACCTACAAGCTGTGCCACCCCGAGGAGCTGGTGCTGCTCGGACACTCT
CTGGGCATCCCCTGGGCTCCCCTGAGCAGCTGCCCCAGCCAGGCCCTGCA
GCTGGCAGGCTGCTTGAGCCAACTCCATAGCGGCCTTTTCCTCTACCAGG
GGCTCCTGCAGGCCCTGGAAGGGATCTCCCCCGAGTTGGGTCCCACCTTG
GACACACTGCAGCTGGACGTCGCCGACTTTGCCACCACCATCTGGCAGCA
GATGGAAGAACTGGGAATGGCCCCTGCCCTGCAGCCCACCCAGGGTGCC
ATGCCGGCCTTCGCCTCTGCTTTCCAGCGCCGGGCAGGAGGGGTCCTGGT
TGCCTCCCATCTGCAGAGCTTCCTGGAGGTGTCGTACCGCGTTCTACGCC
ACCTTGCCCAGCCCTGA
AGCGCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCC
TTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCGGC
CGCTCGAGCATGCATCTAGA
6594Optimized sequence; containing T7 polymerase site,
AfeI and Xba restriction site
TAATACGACTCACTATA
GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACC
ATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCCCTGCAGTT
GCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAGCGACTCCTCTCG
GACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTGAAGTGTCTGGAGCAG
GTGCGAAAGATTCAGGGCGATGGAGCCGCACTCCAAGAGAAGCTCTGCG
CGACATACAAACTTTGCCATCCCGAGGAGCTCGTACTGCTCGGGCACAGC
TTGGGGATTCCCTGGGCTCCTCTCTCGTCCTGTCCGTCGCAGGCTTTGCAG
TTGGCAGGGTGCCTTTCCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGA
CTGCTGCAAGCCCTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGA
CACGTTGCAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGA
TGGAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGCAAT
GCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGTCCTCGTAG
CGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGGGTGCTGAGACAT
CTTGCGCAGCCGTGA
AGCGCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCC
TTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCGGC
CGCTCGAGCATGCATCTAGA
6595mRNA sequence (transcribed)
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCCACC
AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGGCCCUGCAG
UUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAGCGACUCCU
CUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUUUUGAAGUGUCUG
GAGCAGGUGCGAAAGAUUCAGGGCGAUGGAGCCGCACUCCAAGAGAAG
CUCUGCGCGACAUACAAACUUUGCCAUCCCGAGGAGCUCGUACUGCUC
GGGCACAGCUUGGGGAUUCCCUGGGCUCCUCUCUCGUCCUGUCCGUCG
CAGGCUUUGCAGUUGGCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUG
UUCUUGUAUCAGGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAA
UUGGGCCCGACGCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCA
ACAACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCUG
CAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUCAGCGC
AGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCAUUUUUGGAA
GUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCGUGA
AGCGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCCCUUCUUCUCUC
CCUUGCACCUGUACCUCUUGGUCUUUGAAUAAAGCCUGAGUAGGAAG

PCR procedures for the preparation of cDNA are performed using 2×KAPA HIF1™ HotStart ReadyMix by Kapa Biosystems (Woburn, Mass.). This system includes 2×KAPA ReadyMix 12.5 μl; Forward Primer (10 uM) 0.75 μl; Reverse Primer (10 uM) 0.75 μl; Template cDNA 100 ng; and dH2O diluted to 25.0 μl. The reaction conditions are at 95° C. for 5 min. and 25 cycles of 98° C. for 20 sec, then 58° C. for 15 sec, then 72° C. for 45 sec, then 72° C. for 5 min. then 4° C. to termination.

The reverse primer of the instant invention incorporates a poly-T120 for a poly-A120 in the mRNA. Other reverse primers with longer or shorter poly(T) tracts can be used to adjust the length of the poly(A) tail in the mRNA.

The reaction is cleaned up using Invitrogen's PURELINK™ PCR Micro Kit (Carlsbad, Calif.) per manufacturer's instructions (up to 5 μg). Larger reactions will require a cleanup using a product with a larger capacity. Following the cleanup, the cDNA is quantified using the NanoDrop and analyzed by agarose gel electrophoresis to confirm the cDNA is the expected size. The cDNA is then submitted for sequencing analysis before proceeding to the in vitro transcription reaction.

The in vitro transcription reaction generates mRNA containing modified nucleotides or modified RNA. The input nucleotide triphosphate (NTP) mix is made in-house using natural and un-natural NTPs.

A typical in vitro transcription reaction includes the following:

Template cDNA1.0 μg
10x transcription buffer (400 mM2.0 μl
Tris-HCl pH 8.0, 190 mM MgCl2,
50 mM
DTT, 10 mM Spermidine)
Custom NTPs (25 mM each)7.2 μl
RNase Inhibitor 20 U
T7 RNA polymerase3000 U
dH20Up to 20.0 μl. and
Incubation at 37° C. for 3 hr-5 hrs.

The crude IVT mix may be stored at 4° C. overnight for cleanup the next day. 1 U of RNase-free DNase is then used to digest the original template. After 15 minutes of incubation at 37° C., the mRNA is purified using Ambion's MEGACLEAR™ Kit (Austin, Tex.) following the manufacturer's instructions. This kit can purify up to 500 μg of RNA. Following the cleanup, the RNA is quantified using the NanoDrop and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no degradation of the RNA has occurred.

Capping of the mRNA is performed as follows where the mixture includes: IVT RNA 60 μg-180 μg and dH2O up to 72 μl. The mixture is incubated at 65° C. for 5 minutes to denature RNA, and then is transferred immediately to ice.

The protocol then involves the mixing of 10× Capping Buffer (0.5 M Tris-HCl (pH 8.0), 60 mM KCl, 12.5 mM MgCl2) (10.0 μl); 20 mM GTP (5.0 μl); 20 mM S-Adenosyl Methionine (2.5 μl); RNase Inhibitor (100 U); 2′-O-Methyltransferase (400 U); Vaccinia capping enzyme (Guanylyl transferase) (40 U); dH2O (Up to 28 μl); and incubation at 37° C. for 30 minutes for 60 μg RNA or up to 2 hours for 180 μg of RNA.

The mRNA is then purified using Ambion's MEGACLEAR™ Kit (Austin, Tex.) following the manufacturer's instructions. Following the cleanup, the RNA is quantified using the NANODROP™ (ThermoFisher, Waltham, Mass.) and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no degradation of the RNA has occurred. The RNA product may also be sequenced by running a reverse-transcription-PCR to generate the cDNA for sequencing.

Without a poly-T in the cDNA, a poly-A tailing reaction must be performed before cleaning the final product. This is done by mixing Capped IVT RNA (100 μl); RNase Inhibitor (20 U); 10× Tailing Buffer (0.5 M Tris-HCl (pH 8.0), 2.5 M NaCl, 100 mM MgCl2)(12.0 μl); 20 mM ATP (6.0 μl); Poly-A Polymerase (20 U); dH2O up to 123.5 μl and incubation at 37° C. for 30 min. If the poly-A tail is already in the transcript, then the tailing reaction may be skipped and proceed directly to cleanup with Ambion's MEGACLEAR™ kit (Austin, Tex.) (up to 500 μs). Poly-A Polymerase is preferably a recombinant enzyme expressed in yeast.

For studies performed and described herein, the poly-A tail is encoded in the IVT template to comprise 160 nucleotides in length. However, it should be understood that the processivity or integrity of the polyA tailing reaction may not always result in exactly 160 nucleotides. Hence polyA tails of approximately 160 nucleotides, e.g, about 150-165, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164 or 165 are within the scope of the invention.

5′-capping of modified RNA may be completed concomitantly during the in vitro-transcription reaction using the following chemical RNA cap analogs to generate the 5′-guanosine cap structure according to manufacturer protocols: 3′-O-Me-m7G(5′)ppp(5′) G [the ARCA cap]; G(5)ppp(5′)A; G(5′)ppp(5′)G; m7G(5′)ppp(5′)A; m7G(5′)ppp(5′)G (New England BioLabs, Ipswich, Mass.). 5′-capping of modified RNA may be completed post-transcriptionally using a Vaccinia Virus Capping Enzyme to generate the “Cap 0” structure: m7G(5′)ppp(5′)G (New England BioLabs, Ipswich, Mass.). Cap 1 structure may be generated using both Vaccinia Virus Capping Enzyme and a 2′-O methyl-transferase to generate: m7G(5′)ppp(5′)G-2′-O-methyl. Cap 2 structure may be generated from the Cap 1 structure followed by the 2′-O-methylation of the 5′-antepenultimate nucleotide using a 2′-O methyl-transferase. Cap 3 structure may be generated from the Cap 2 structure followed by the 2′-O-methylation of the 5′-preantepenultimate nucleotide using a 2′-O methyl-transferase. Enzymes are preferably derived from a recombinant source.

When transfected into mammalian cells, the modified mRNAs have a stability of between 12-18 hours or more than 18 hours, e.g., 24, 36, 48, 60, 72 or greater than 72 hours.

A. Protein Expression Assay

Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 6567; mRNA sequence fully modified with 5-methylcytidine at each cytidine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6570 with a polyA tail approximately 160 nucleotides in length not shown in sequence) containing the ARCA (3′ O-Me-m7G(5)ppp(5′)G) cap analog or the Cap1 structure can be transfected into human primary keratinocytes at equal concentrations. 6, 12, 24 and 36 hours post-transfection the amount of G-CSF secreted into the culture medium can be assayed by ELISA. Synthetic mRNAs that secrete higher levels of G-CSF into the medium would correspond to a synthetic mRNA with a higher translationally-competent Cap structure.

B. Purity Analysis Synthesis

Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 6567; mRNA sequence fully modified with 5-methylcytidine at each cytidine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6570 with a polyA tail approximately 160 nucleotides in length not shown in sequence) containing the ARCA cap analog or the Cap1 structure crude synthesis products can be compared for purity using denaturing Agarose-Urea gel electrophoresis or HPLC analysis. Synthetic mRNAs with a single, consolidated band by electrophoresis correspond to the higher purity product compared to a synthetic mRNA with multiple bands or streaking bands. Synthetic mRNAs with a single HPLC peak would also correspond to a higher purity product. The capping reaction with a higher efficiency would provide a more pure mRNA population.

C. Cytokine Analysis

Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 6567; mRNA sequence fully modified with 5-methylcytidine at each cytidine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6570 with a polyA tail approximately 160 nucleotides in length not shown in sequence) containing the ARCA cap analog or the Cap 1 structure can be transfected into human primary keratinocytes at multiple concentrations. 6, 12, 24 and 36 hours post-transfection the amount of pro-inflammatory cytokines such as TNF-alpha and IFN-beta secreted into the culture medium can be assayed by ELISA. Synthetic mRNAs that secrete higher levels of pro-inflammatory cytokines into the medium would correspond to a synthetic mRNA containing an immune-activating cap structure.

D. Capping Reaction Efficiency

Synthetic mRNAs encoding human G-CSF (cDNA shown in SEQ ID NO: 6567; mRNA sequence fully modified with 5-methylcytidine at each cytidine and pseudouridine replacement at each uridine site shown in SEQ ID NO: 6570 with a polyA tail approximately 160 nucleotides in length not shown in sequence) containing the ARCA cap analog or the Cap 1 structure can be analyzed for capping reaction efficiency by LC-MS after capped mRNA nuclease treatment. Nuclease treatment of capped mRNAs would yield a mixture of free nucleotides and the capped 5′-5-triphosphate cap structure detectable by LC-MS. The amount of capped product on the LC-MS spectra can be expressed as a percent of total mRNA from the reaction and would correspond to capping reaction efficiency. The cap structure with higher capping reaction efficiency would have a higher amount of capped product by LC-MS.

Individual modified RNAs (200-400 ng in a 20 μl volume) or reverse transcribed PCR products (200-400 ng) are loaded into a well on a non-denaturing 1.2% Agarose E-Gel (Invitrogen, Carlsbad, Calif.) and run for 12-15 minutes according to the manufacturer protocol.

Modified RNAs in TE buffer (1 μl) are used for Nanodrop UV absorbance readings to quantitate the yield of each modified RNA from an in vitro transcription reaction.

Signal-sensor polynucleotides may be formulated for in vitro and in vivo experiments according to the methods taught in International Application PCT/US12/069610 filed Dec. 14, 2012, the contents of which are incorporated herein by reference in their entirety.

Signal-sensor polynucleotides may be investigated using the methods described in co-pending International Patent application No. PCT/US2013/030070 filed Mar. 9, 2013 and U.S. Patent Application No. 61/681,742 filed Aug. 10, 2012 (MNC2), the contents of which are incorporated herein by reference in their entirety.

Signal-sensor polynucleotides may be investigated in any number of cancer or normal cell lines. Cell lines useful in the present invention include those from ATCC (Manassas, Va.) and are listed in Table 13.

TABLE 13
Cell lines
ATCC
NumberHybridoma or Cell line DescriptionName
CCL-171hom*o sapiens (human) Source: Organ: lungMRC-5
Disease: normal
Cell Type: fibroblast
CCL-185hom*o sapiens (human) Source: Organ: lungA549
Disease: carcinoma
CCL-248hom*o sapiens (human) Source: Organ: colonT84
Disease: colorectal carcinoma
Derived from metastatic site: lung
CCL-256hom*o sapiens (human) Source: Organ: lungNCI-H2126
Disease: adenocarcinoma; non-small cell lung cancer[H2126]
Derived from metastatic site: pleural effusion
CCL-257hom*o sapiens (human) Source: Organ: lungNCI-H1688
Disease: carcinoma; classic small cell lung cancer[H1688]
CCL-75hom*o sapiens (human) Source: Organ: lungWI-38
Disease: normal
Cell Type: fibroblast
CCL-75.1hom*o sapiens (human) Source: Organ: lungWI-38 VA-13
Cell Type: fibroblastSV40 transformedsubline 2RA
CCL-95.1hom*o sapiens (human) Source: Organ: lungWI-26 VA4
Cell Type: SV40 transformed
CRL-10741hom*o sapiens (human) Source: Organ: liverC3A [HepG2/C3A,
Disease: hepatocellular carcinomaderivative of Hep
G2 (ATCC HB-
8065)]
CRL-11233hom*o sapiens (human) Source: Organ: liverTHLE-3
Tissue: left lobe
Cell Type: epithelialimmortalized with SV40 large T
antigen
CRL-11351hom*o sapiens (human) Source: Organ: lungH69AR
Disease: carcinoma; small cell lung cancer; multidrug
resistant
Cell Type: epithelial
CRL-1848hom*o sapiens (human) Source: Organ: lungNCI-H292 [H292]
Disease: mucoepidermoid pulmonary carcinoma
CRL-1918hom*o sapiens (human) Source: Organ: pancreasCFPAC-1
Disease: ductal adenocarcinoma; cystic fibrosis
Derived from metastatic site: liver metastasis
CRL-1973hom*o sapiens (human) Source: Organ: testisNTERA-2 cl.D1
Disease: malignant pluripotent embryonal carcinoma[NT2/D1]
Derived from metastatic site: lung
CRL-2049hom*o sapiens (human) Source: Organ: lungDMS 79
Disease: carcinoma; small cell lung cancer
CRL-2062hom*o sapiens (human) Source: Organ: lungDMS 53
Disease: carcinoma; small cell lung cancer
CRL-2064hom*o sapiens (human) Source: Organ: lungDMS 153
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: liver
CRL-2066hom*o sapiens (human) Source: Organ: lungDMS 114
Disease: carcinoma; small cell lung cancer
CRL-2081hom*o sapiens (human) Source: Disease: biphasicMSTO-211H
mesothelioma
Derived from metastatic site: lung
CRL-2170hom*o sapiens (human) Source: Organ: lungSW 1573 [SW-
Disease: alveolar cell carcinoma1573, SW1573]
CRL-2177hom*o sapiens (human) Source: Organ: lungSW 1271 [SW-
Disease: carcinoma; small cell lung cancer1271, SW1271]
CRL-2195hom*o sapiens (human) Source: Organ: lungSHP-77
Disease: carcinoma; small cell lung cancer
Cell Type: large cell, variant;
CRL-2233hom*o sapiens (human) Source: Organ: liverSNU-398
Disease: hepatocellular carcinoma
CRL-2234hom*o sapiens (human) Source: Organ: liverSNU-449
Tumor Stage: grade II-III/IV
Disease: hepatocellular carcinoma
CRL-2235hom*o sapiens (human) Source: Organ: liverSNU-182
Tumor Stage: grade III/IV
Disease: hepatocellular carcinoma
CRL-2236hom*o sapiens (human) Source: Organ: liverSNU-475
Tumor Stage: grade II-IV/V
Disease: hepatocellular carcinoma
CRL-2237hom*o sapiens (human) Source: Organ: liverSNU-387
Tumor Stage: grade IV/V
Disease: pleomorphic hepatocellular carcinoma
CRL-2238hom*o sapiens (human) Source: Organ: liverSNU-423
Tumor Stage: grade III/IV
Disease: pleomorphic hepatocellular carcinoma
CRL-2503hom*o sapiens (human) Source: Organ: lungNL20
Tissue: bronchus
Disease: normal
CRL-2504hom*o sapiens (human) Source: Organ: lungNL20-TA [NL20T-
Tissue: bronchusA]
Disease: normal
CRL-2706hom*o sapiens (human) Source: Organ: liverTHLE-2
Tissue: left lobe
Cell Type: epithelialSV40 transformed
CRL-2741hom*o sapiens (human) Source: Organ: lungHBE135-E6E7
Tissue: bronchus
Cell Type: epithelialHPV-16 E6/E7 transformed
CRL-2868hom*o sapiens (human) Source: Organ: lungHCC827
Disease: adenocarcinoma
Cell Type: epithelial
CRL-2871hom*o sapiens (human) Source: Organ: lungHCC4006
Disease: adenocarcinoma
Derived from metastatic site: pleural effusion
Cell Type: epithelial
CRL-5800hom*o sapiens (human) Source: Organ: lungNCI-H23 [H23]
Disease: adenocarcinoma; non-small cell lung cancer
CRL-5803hom*o sapiens (human) Source: Organ: lungNCI-H1299
Disease: carcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5804hom*o sapiens (human) Source: Organ: lungNCI-H187 [H187]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5807hom*o sapiens (human) Source: Organ: lungNCI-H358 [H-358,
Tissue: bronchiole; alveolusH358]
Disease: bronchioalveolar carcinoma; non-small cell
lung cancer
CRL-5808hom*o sapiens (human) Source: Organ: lungNCI-H378 [H378]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5810hom*o sapiens (human) Source: Organ: lungNCI-H522 [H522]
Tumor Stage: stage 2
Disease: adenocarcinoma; non-small cell lung cancer
CRL-5811hom*o sapiens (human) Source: Organ: lungNCI-H526 [H526]
Tumor Stage: stage E
Disease: carcinoma; variant small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5815hom*o sapiens (human) Source: Organ: lungNCI-H727 [H727]
Tissue: bronchus
Disease: carcinoid
CRL-5816hom*o sapiens (human) Source: Organ: lungNCI-H810 [H810]
Tumor Stage: stage 2
Disease: carcinoma; non-small cell lung cancer
CRL-5817hom*o sapiens (human) Source: Organ: lungNCI-H889 [H889]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5818hom*o sapiens (human) Source: Organ: lungNCI-H1155
Disease: carcinoma; non-small cell lung cancer[H1155]
Derived from metastatic site: lymph node
CRL-5819hom*o sapiens (human) Source: Organ: lungNCI-H1404
Disease: papillary adenocarcinoma[H1404]
Derived from metastatic site: lymph node
CRL-5822hom*o sapiens (human) Source: Organ: stomachNCI-N87 [N87]
Disease: gastric carcinoma
Derived from metastatic site: liver
CRL-5823hom*o sapiens (human) Source: Organ: lungNCI-H196 [H196]
Tumor Stage: stage E
Disease: carcinoma; variant small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5824hom*o sapiens (human) Source: Organ: lungNCI-H211 [H211]
Tumor Stage: stage E
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5825hom*o sapiens (human) Source: Organ: lungNCI-H220 [H220]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5828hom*o sapiens (human) Source: Organ: lungNCI-H250 [H250]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: brain
CRL-5831hom*o sapiens (human) Source: Organ: lungNCI-H524 [H524]
Tumor Stage: stage L
Disease: carcinoma; variant small cell lung cancer
Derived from metastatic site: lymph node
CRL-5834hom*o sapiens (human) Source: Organ: lungNCI-H647 [H647]
Tumor Stage: stage 3A
Disease: adenosquamous carcinoma; non-small cell lung
cancer
Derived from metastatic site: pleural effusion
CRL-5835hom*o sapiens (human) Source: Organ: lungNCI-H650 [H650]
Disease: bronchioalveolar carcinoma; non-small cell
lung cancer
Derived from metastatic site: lymph node
CRL-5836hom*o sapiens (human) Source: Organ: lungNCI-H711 [H711]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5837hom*o sapiens (human) Source: Organ: lungNCI-H719 [H719]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5840hom*o sapiens (human) Source: Organ: lungNCI-H740 [H740]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5841hom*o sapiens (human) Source: Organ: lungNCI-H748 [H748]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5842hom*o sapiens (human) Source: Organ: lungNCI-H774 [H774]
Tumor Stage: stage E
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: soft tissue
CRL-5844hom*o sapiens (human) Source: Organ: lungNCI-H838 [H838]
Tumor stage: 3B
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5845hom*o sapiens (human) Source: Organ: lungNCI-H841 [H841]
Tumor Stage: stage L
Disease: carcinoma; variant small cell lung cancer
Derived from metastatic site: lymph node
CRL-5846hom*o sapiens (human) Source: Organ: lungNCI-H847 [H847]
Tumor Stage: stage L
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5849hom*o sapiens (human) Source: Organ: lungNCI-H865 [H865]
Tumor Stage: stage L
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5850hom*o sapiens (human) Source: Organ: lungNCI-H920 [H920]
Tumor Stage: stage 4
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5853hom*o sapiens (human) Source: Organ: lungNCI-H1048
Disease: carcinoma; small cell lung cancer[H1048]
Derived from metastatic site: pleural effusion
CRL-5855hom*o sapiens (human) Source: Organ: lungNCI-H1092
Tumor Stage: stage E[H1092]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5856hom*o sapiens (human) Source: Organ: lungNCI-H1105
Tumor Stage: stage E[H1105]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5858hom*o sapiens (human) Source: Organ: lungNCI-H1184
Tumor Stage: stage L[H1184]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: lymph node
CRL-5859hom*o sapiens (human) Source: Organ: lungNCI-H1238
Tumor Stage: stage E[H1238]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5864hom*o sapiens (human) Source: Organ: lungNCI-H1341
Disease: carcinoma; small cell lung cancer[H1341]
Derived from metastatic site: cervix
CRL-5867hom*o sapiens (human) Source: Organ: lungNCI-H1385
Tumor Stage: stage 3A[H1385]
Disease: carcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5869hom*o sapiens (human) Source: Organ: lungNCI-H1417
Tumor Stage: stage E[H1417]
Disease: carcinoma; classic small cell lung cancer
CRL-5870hom*o sapiens (human) Source: Organ: lungNCI-H1435
Disease: adenocarcinoma; non-small cell lung cancer[H1435]
CRL-5871hom*o sapiens (human) Source: Organ: lungNCI-H1436
Tumor Stage: stage E[H1436]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5872hom*o sapiens (human) Source: Organ: lungNCI-H1437
Tumor Stage: stage 1[H1437]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5874hom*o sapiens (human) Source: Organ: lungNCI-H1522
Tumor Stage: stage E[H1522]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5875hom*o sapiens (human) Source: Organ: lungNCI-H1563
Disease: adenocarcinoma; non-small cell lung cancer[H1563]
CRL-5876hom*o sapiens (human) Source: Organ: lungNCI-H1568
Disease: adenocarcinoma; non-small cell lung cancer[H1568]
Derived from metastatic site: lymph node
CRL-5877hom*o sapiens (human) Source: Organ: lungNCI-H1573
Tumor Stage: stage 4[H1573]
Disease: adenocarcinoma
Derived from metastatic site: soft tissue
CRL-5878hom*o sapiens (human) Source: Organ: lungNCI-H1581
Tumor Stage: stage 4[H1581]
Disease: non-small cell lung cancer
Cell Type: large cell;
CRL-5879hom*o sapiens (human) Source: Tumor Stage: stage ENCI-H1618
Disease: carcinoma; small cell lung cancer[H1618]
Derived from metastatic site: bone marrow
CRL-5881hom*o sapiens (human) Source: Organ: lungNCI-H1623
Tumor Stage: stage 3B[H1623]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5883hom*o sapiens (human) Source: Organ: lungNCI-H1650 [H-
Tumor Stage: stage 3B1650, H1650]
Disease: adenocarcinoma; bronchoalveolar carcinoma
Derived from metastatic site: pleural effusion
CRL-5884hom*o sapiens (human) Source: Organ: lungNCI-H1651
Disease: adenocarcinoma; non-small cell lung cancer[H1651]
CRL-5885hom*o sapiens (human) Source: Organ: lungNCI-H1666 [H-
Disease: adenocarcinoma; bronchoalveolar carcinoma1666, H1666]
Derived from metastatic site: pleural effusion
CRL-5886hom*o sapiens (human) Source: Organ: lungNCI-H1672
Tumor Stage: stage L[H1672]
Disease: carcinoma; classic small cell lung cancer
CRL-5887hom*o sapiens (human) Source: Organ: lungNCI-H1693
Tumor Stage: stage 3B[H1693]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5888hom*o sapiens (human) Source: Organ: lungNCI-H1694
Tumor Stage: stage E[H1694]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: ascites
CRL-5889hom*o sapiens (human) Source: Organ: lungNCI-H1703
Tumor Stage: stage 1[H1703]
Disease: non-small cell lung cancer
Cell Type: squamous cell;
CRL-5891hom*o sapiens (human) Source: Organ: lungNCI-H1734 [H-
Disease: adenocarcinoma; non-small cell lung cancer1734, H1734]
CRL-5892hom*o sapiens (human) Source: Organ: lungNCI-H1755
Tumor Stage: stage 4[H1755]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: liver
CRL-5892hom*o sapiens (human) Source: Organ: lungNCI-H1755
Tumor Stage: stage 4[H1755]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: liver
CRL-5893hom*o sapiens (human) Source: Organ: lungNCI-H1770
Tumor Stage: stage 4[H1770]
Disease: carcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
Cell Type: neuroendocrine;
CRL-5896hom*o sapiens (human) Source: Organ: lungNCI-H1793
Disease: adenocarcinoma; non-small cell lung cancer[H1793]
CRL-5898hom*o sapiens (human) Source: Organ: lungNCI-H1836
Tumor Stage: stage L[H1836]
Disease: carcinoma; classic small cell lung cancer
CRL-5899hom*o sapiens (human) Source: Organ: lungNCI-H1838
Disease: adenocarcinoma; non-small cell lung cancer[H1838]
CRL-5900hom*o sapiens (human) Source: Organ: lungNCI-H1869
Tumor Stage: stage 4[H1869]
Disease: non-small cell lung cancer
Derived from metastatic site: pleural effusion
Cell Type: squamous cell;
CRL-5902hom*o sapiens (human) Source: Organ: lungNCI-H1876
Tumor Stage: stage E[H1876]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5903hom*o sapiens (human) Source: Organ: lungNCI-H1882
Tumor Stage: stage E[H1882]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5904hom*o sapiens (human) Source: Organ: lungNCI-H1915
Tumor Stage: stage 4[H1915]
Disease: poorly differentiated carcinoma; non-small cell
lung cancer
Derived from metastatic site: brain
Cell Type: large cell;
CRL-5906hom*o sapiens (human) Source: Organ: lungNCI-H1930
Tumor Stage: stage L[H1930]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: lymph node
CRL-5907hom*o sapiens (human) Source: Organ: lungNCI-H1944
Tumor Stage: stage 3B[H1944]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: soft tissue
CRL-5908hom*o sapiens (human) Source: Organ: lungNCI-H1975 [H-
Disease: adenocarcinoma; non-small cell lung cancer1975, H1975]
CRL-5909hom*o sapiens (human) Source: Organ: lungNCI-H1993
Tumor Stage: stage 3A[H1993]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5912hom*o sapiens (human) Source: Organ: lungNCI-H2023
Tumor Stage: stage 3A[H2023]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5913hom*o sapiens (human) Source: Organ: lungNCI-H2029
Tumor Stage: stage E[H2029]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: lymph node
CRL-5914hom*o sapiens (human) Source: Organ: lungNCI-H2030
Disease: adenocarcinoma; non-small cell lung cancer[H2030]
Derived from metastatic site: lymph node
CRL-5917hom*o sapiens (human) Source: Organ: lungNCI-H2066
Tumor Stage: stage 1[H2066]
Disease: mixed; small cell lung cancer; adenocarcinoma;
squamous cell carcinoma
CRL-5918hom*o sapiens (human) Source: Organ: lungNCI-H2073
Tumor Stage: stage 3A[H2073]
Disease: adenocarcinoma; non-small cell lung cancer
CRL-5920hom*o sapiens (human) Source: Organ: lungNCI-H2081
Tumor Stage: stage E[H2081]
Disease: carcinoma; classic small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5921hom*o sapiens (human) Source: Organ: lungNCI-H2085
Disease: adenocarcinoma; non-small cell lung cancer[H2085]
CRL-5922hom*o sapiens (human) Source: Organ: lungNCI-H2087
Tumor Stage: stage 1[H2087]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5923hom*o sapiens (human) Source: Organ: lungNCI-H2106
Tissue: neuroendocrine[H2106]
Tumor Stage: stage 4
Disease: non-small cell lung cancer
Derived from metastatic site: lymph node
CRL-5924hom*o sapiens (human) Source: Organ: lungNCI-H2110
Disease: non-small cell lung cancer[H2110]
Derived from metastatic site: pleural effusion
CRL-5926hom*o sapiens (human) Source: Organ: lungNCI-H2135
Disease: non-small cell lung cancer[H2135]
CRL-5927hom*o sapiens (human) Source: Organ: lungNCI-H2141
Tumor Stage: stage E[H2141]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: lymph node
CRL-5929hom*o sapiens (human) Source: Organ: lungNCI-H2171
Tumor Stage: stage E[H2171]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-5930hom*o sapiens (human) Source: Organ: lungNCI-H2172
Disease: non-small cell lung cancer[H2172]
CRL-5931hom*o sapiens (human) Source: Organ: lungNCI-H2195
Tumor Stage: stage E[H2195]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5932hom*o sapiens (human) Source: Organ: lungNCI-H2196
Tumor Stage: stage E[H2196]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5933hom*o sapiens (human) Source: Organ: lungNCI-H2198
Tumor Stage: stage E[H2198]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: lymph node
CRL-5934hom*o sapiens (human) Source: Organ: lungNCI-H2227
Tumor Stage: stage E[H2227]
Disease: carcinoma; small cell lung cancer
CRL-5935hom*o sapiens (human) Source: Organ: lungNCI-H2228
Disease: adenocarcinoma; non-small cell lung cancer[H2228]
CRL-5938hom*o sapiens (human) Source: Organ: lungNCI-H2286
Tumor Stage: stage 1[H2286]
Disease: mixed; small cell lung cancer; adenocarcinoma;
squamous cell carcinoma
CRL-5939hom*o sapiens (human) Source: Organ: lungNCI-H2291
Disease: adenocarcinoma; non-small cell lung cancer[H2291]
Derived from metastatic site: lymph node
CRL-5940hom*o sapiens (human) Source: Organ: lungNCI-H2330
Tumor Stage: stage L[H2330]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: lymph node
CRL-5941hom*o sapiens (human) Source: Organ: lungNCI-H2342
Tumor Stage: stage 3A[H2342]
Disease: adenocarcinoma; non-small cell lung cancer
CRL-5942hom*o sapiens (human) Source: Organ: lungNCI-H2347
Tumor Stage: stage 1[H2347]
Disease: adenocarcinoma; non-small cell lung cancer
CRL-5944hom*o sapiens (human) Source: Organ: lungNCI-H2405
Tumor Stage: stage 4[H2405]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: ascites
CRL-5945hom*o sapiens (human) Source: Organ: lungNCI-H2444
Disease: non-small cell lung cancer[H2444]
CRL-5975hom*o sapiens (human) Source: Organ: lungUMC-11
Disease: carcinoid
CRL-5976hom*o sapiens (human) Source: Organ: lungNCI-H64 [H64]
Tumor Stage: stage E
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: lymph node
CRL-5978hom*o sapiens (human) Source: Organ: lungNCI-H735 [H735]
Tumor Stage: stage E
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: liver
CRL-5978hom*o sapiens (human) Source: Organ: lungNCI-H735 [H735]
Tumor Stage: stage E
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: liver
CRL-5982hom*o sapiens (human) Source: Organ: lungNCI-H1963
Tumor Stage: stage L[H1963]
Disease: carcinoma; small cell lung cancer
CRL-5983hom*o sapiens (human) Source: Organ: lungNCI-H2107
Tumor Stage: stage E[H2107]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5984hom*o sapiens (human) Source: Organ: lungNCI-H2108
Tumor Stage: stage E[H2108]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
CRL-5985hom*o sapiens (human) Source: Organ: lungNCI-H2122
Tumor Stage: stage 4[H2122]
Disease: adenocarcinoma; non-small cell lung cancer
Derived from metastatic site: pleural effusion
CRL-7343hom*o sapiens (human) Source: Organ: lungHs 573.T
Disease: cancer
CRL-7344hom*o sapiens (human) Source: Organ: lungHs 573.Lu
CRL-8024hom*o sapiens (human) Source: Organ: liverPLC/PRF/5
Disease: hepatoma
Cell Type: Alexander cells;
CRL-9609hom*o sapiens (human) Source: Organ: lungBEAS-2B
Tissue: bronchus
Disease: normal
Cell Type: epithelialvirus transformed
HB-8065hom*o sapiens (human) Source: Organ: liverHep G2
Disease: hepatocellular carcinoma
HTB-105hom*o sapiens (human) Source: Organ: testesTera-1
Disease: embryonal carcinoma, malignant
Derived from metastatic site: lung
HTB-106hom*o sapiens (human) Source: Disease: malignantTera-2
embryonal carcinoma
Derived from metastatic site: lung
HTB-119hom*o sapiens (human) Source: Organ: lungNCI-H69 [H69]
Disease: carcinoma; small cell lung cancer
HTB-120hom*o sapiens (human) Source: Organ: lungNCI-H128 [H128]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: pleural effusion
HTB-168hom*o sapiens (human) Source: Organ: lungChaGo-K-1
Tissue: bronchus
Disease: bronchogenic carcinoma
HTB-171hom*o sapiens (human) Source: Organ: lungNCI-H446 [H446]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: pleural effusion
HTB-172hom*o sapiens (human) Source: Organ: lungNCI-H209 [H209]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
HTB-173hom*o sapiens (human) Source: Organ: lungNCI-H146 [H146]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
HTB-174hom*o sapiens (human) Source: Organ: lungNCI-H441 [H441]
Disease: papillary adenocarcinoma
HTB-175hom*o sapiens (human) Source: Organ: lungNCI-H82 [H82]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: pleural effusion
HTB-177hom*o sapiens (human) Source: Organ: lungNCI-H460 [H460]
Disease: carcinoma; large cell lung cancer
Derived from metastatic site: pleural effusion
HTB-178hom*o sapiens (human) Source: Organ: lungNCI-H596 [H596]
Disease: adenosquamous carcinoma
HTB-179hom*o sapiens (human) Source: Organ: lungNCI-H676B
Disease: adenocarcinoma[H676B]
Derived from metastatic site: pleural effusion
HTB-180hom*o sapiens (human) Source: Organ: lungNCI-H345 [H345]
Disease: carcinoma; small cell lung cancer
Derived from metastatic site: bone marrow
HTB-181hom*o sapiens (human) Source: Organ: lungNCI-H820 [H820]
Disease: papillary adenocarcinoma
Derived from metastatic site: lymph node
HTB-182hom*o sapiens (human) Source: Organ: lungNCI-H520 [H520]
Disease: squamous cell carcinoma
HTB-183hom*o sapiens (human) Source: Organ: lungNCI-H661 [H661]
Disease: carcinoma; large cell lung cancer
Derived from metastatic site: lymph node
HTB-184hom*o sapiens (human) Source: Organ: lungNCI-H510A
Disease: carcinoma; small cell lung cancer;[H510A, NCI-
extrapulmonary originH510]
Derived from metastatic site: adrenal gland
HTB-52hom*o sapiens (human) Source: Organ: liverSK-HEP-1
Tissue: ascites
Disease: adenocarcinoma
HTB-53hom*o sapiens (human) Source: Organ: lungA-427
Disease: carcinoma
HTB-54hom*o sapiens (human) Source: Organ: lungCalu-1
Tumor Stage: grade III
Disease: epidermoid carcinoma
Derived from metastatic site: pleura
HTB-55hom*o sapiens (human) Source: Organ: lungCalu-3
Disease: adenocarcinoma
Derived from metastatic site: pleural effusion
HTB-56hom*o sapiens (human) Source: Organ: unknown,Calu-6
probably lung
Disease: anaplastic carcinoma
HTB-57hom*o sapiens (human) Source: Organ: lungSK-LU-1
Disease: adenocarcinoma
HTB-58hom*o sapiens (human) Source: Organ: lungSK-MES-1
Disease: squamous cell carcinoma
Derived from metastatic site: pleural effusion
HTB-59hom*o sapiens (human) Source: Organ: lungSW 900 [SW-900,
Tumor Stage: grade IVSW900]
Disease: squamous cell carcinoma
HTB-64hom*o sapiens (human) Source: Disease: malignantMalme-3M
melanoma
Derived from metastatic site: lung
HTB-79hom*o sapiens (human) Source: Organ: pancreasCapan-1
Disease: adenocarcinoma
Derived from metastatic site: liver

Various animal models are available for the study of the signal-sensor polynucleotides of the present invention. These include, among others, models for lung and liver cancers.

The lung cancer model of f*ckazawa et al (Anticancer Research, 2010; 30: 4193-4200) is employed in studies of signal-sensor polynucleotides. Briefly, a congenic mouse is created by crossing a ubiquitously expressing dominant negative Myc (Omomyc) mouse with a KRAS mutation-positive lung cancer model mouse. In the presence of Omomyc, lung tumors caused by the expression of mutated KRAS regresses in the congenic mouse, indicating that Omomyc caused tumor cell death of KRAS mutation-positive lung cancer.

Human lung cancer xenografts are also prepared by the method of f*ckazawa. Briefly, human lung cancer xenografts are established in 4-week-old female BALB/C nude mice (Charles River Laboratories Japan, Kanagawa, Japan) by subcutaneous inoculation of 4×106 A549 cells into the dorsal flank. The mice are randomly assigned into six groups (n=6/group). After the tumors reach a diameter of about 0.5 cm (approximately 6 days after tumor inoculations), each group of mice are injected with 100 μl solution containing PBS, 5×1010 vp of control or signal-sensor polynucleotide into the dorsal flank tumor for the selected dosing regimen. Animals are then observed closely and survival studies or other analyses are performed.

The LSL-KRASG12D: TRE Omomyc:CMV rtTA triple transgenic model involves the use of an adenovirus expressing Cre recombinase which is administered via inhalation to induce oncogene expression via excision of the floxed STOP codon, and ubiquitous Omomyc expression is controlled via doxycycline. The model is reported in Soucek et al. (Nature, 1-5 (2008)). The mice of Soucek may be crossed with the LSLKRASG12D single transgenic mice (Jackson Laboratories) and may be used for inhalation delivered or otherwise lung-delivered studies of signal-sensor polynucleotides expressing MYC inhibitor D or other oncology related polypeptide.

Mouse-in-mouse models may also be employed. Such models are akin to the p53−/−:c-Myc overexpressing HCC model of Zender (Cell. 2006 Jun. 30; 125(7): 1253-1267).

Design of such models involve starting with the WT or tumor suppressor deleted (such as p53−/−) 129 Sv/Ev Mm ES cell clone; introduction of liver activated protein (LAP) promoter directed tetracycline transactivator (tTA) and tetO-luciferase for liver specific imaging; freezing the resulting LAP-tTA: tetO-luciferase clones to be used for c-Myc as well as other liver relevant programs oncogene; adding tetO driven oncogene, e.g. tetOcMyc; Freeze resulting LAP-tTA: tetO-luciferase: tetO-MYC clones; injecting resulting ES clones into C57Bl/6 blastocytes and implant in pseudo pregnant mothers whereby the resulting chimeric animals are the tumor model upon removal of doxycycline (i.e. Tet-Off). The model will ideally evince inducible nodules of c-Myc-driven, luciferase-expressing HCC surrounded by normal hepatocytes.

Orthotopic HCC models using the HEP3B cell lines in mice may also be used (Crown Bio).

Nongermline genetically engineered mouse model (NGEMM) platform is a platform that could also be utilized for exploring signal-sensor polynucleotides.

Hypoxia-inducible factors (HIFs) control cellular adaptation to oxygen deprivation. Cancer cells engage HIFs to sustain their growth in adverse conditions, thus promoting a cellular reprograming that includes metabolism, proliferation, survival and mobility. HIFs overexpression in human cancer biopsies correlates with high metastasis and mortality.

Hifs regulate genes related to metabolism such as GLUT1, GLUT3, ALDOA, ENOL, GAPDH, HK1, HK2, PFKL, PGK1, PKM2, LDHA, proliferation such as IGF-2, TGFA, VEGFA, survival such as TERT, NANOG, OCT4 and cell migration-invasion such as ZEB1, ZEB2, SNAI2, MMP14, MMP9, AMF, MET, PTHrP. (Keith, et al Nat Rev Cancer 2012; 12:9-22).

To investigate the destabilization of cancer, one or more signal-sensor polynucleotides may be administered to the cancer cell. The selection of the sequence, dose or administrative route is optionally informed by diagnostic evaluation of the cell, tumor, tissue or organism including, but not limited to, expression profiling of the cancer, metabolic evaluation (hypoxic, acidotic), apoptotic vs. survival profiling, cell cycle vs. senescent profiling, immune sensitivities, and/or evaluation of stromal factors.

In one arm of the study signal-sensor polynucleotides encoding either or both oncology related polypeptides, CITED4 and SHARP1 are administered where administration of either or both results in the inhibition of the transcriptome of HIF-1 alpha in cancer cells. Suppression of HIF1-alpha gene regulated expression occurs upon administration with higher suppression when both polynucleotides are administered together. Reporter constructs such as luciferase under HIF1-alpha are used in the manner similar to the methods disclosed in van de Sluis et al, (J Clin Invest. 2010; 120(6):2119-2130). It is known that both CITED4 and SHARP1 expression results in decreased HIF1-alpha and concomitant reduction in HIF1-alpha regulated gene expression. Evaluation of cell death and/or proliferation is also performed.

Additional experiments involve the use a cancer cell line where CITED4 and SHARP1 are themselves down regulated either under hypoxic conditions. Therefore a positive result demonstrates that specifically targeting the metabolic profile (in this case hypoxic-adaptations of CITED4 and SHAPR1) with replacement of native proteins via signal-sensor polynucleotides can directly impact the transcriptome and survival advantage of cancer cells with this profile. Further, the data would show that the relative impact of signal-sensor polynucleotide vs. vehicle under hypoxic conditions was more significant for cancer cells than for normal cells. (i.e., the cancer cells have a disproportionate survival advantage based on their CITED4+SHARP1 down regulation) that makes them more sensitive to the replacement of this protein then a normal cell is to overproduction of it. It is understood that a cancer cell will likely be experiencing hypoxic conditions and that a normal cell under normoxic conditions might be able to tolerate CITED4 and SHARP1 over expression because the normal cell is not dependent on HIF1alpha transcriptome for survival advantage.

In vivo experiments are performed according to the design of the in vitro experiments where the animal model is one evincing metastasis in the cancer setting because HIF-1 alpha appears to confer the largest portion of its advantage in metastasis. Animals are administered the signal-sensor polynucleotide compared to no treatment or a control polynucleotide. Animal cells, tissues and/or organs are then evaluated for alterations in gene expression profiles or transcriptome levels.

Two nuclear export signals (NES) which may be incorporated into the Signal-sensor polynucleotides of the present invention include those reported by Muller, et al (Traffic, 2009, 10: 514-527) and are associated with signaling via the gene COMMD1. These are NES1, PVAIIELEL (SEQ ID NO 6596) and NES2, VNQILKTLSE (SEQ ID NO 6597).

Nuclear localization signals may also be used. One such sequence is PKKKRKV (SEQ ID NO: 6598).

Cell lines or mice are administered one or more signal-sensor polynucleotides having a NLS or NES encoded therein. Upon administration the polynucleotide is trafficked to an alternate location, e.g., into the nucleus using the NLS. The oncology related polypeptide having the NLS would be trafficked to the nucleus where it would deliver either a survival or death signal to the nuclear microenvironment. Polypeptides which may be localized to the nucleus include those with altered binding properties for DNA which will function to alter the expression profile of the cell in a therapeutically beneficial manner for the cell, tissue or organism.

In one experiment, the signal-sensor polynucleotide encodes a COMMD1 mut1/mut2+NLS (e.g., both NES signals disrupted plus a NLS added) following the methods of Muller et al, (Traffic 2009; 10: 514-527) and van de Sluis et al, (J Clin Invest. 2010; 120 (6):2119-2130). The signal sequence may encode an oncology related polypeptide or a scrambled sequence which is not translatable. The signal sequence encoded would interact with HIF1-alpha to alter the transcriptome of the cancer cells.

The experiment is repeated under normal and hypoxic conditions.

Once identified the HIF1-alpha dependent signal-sensor polynucleotide is tested in cancer cell lines clonal survival or a marker of apoptosis is measured and compared to control or mock treated cells.

Using the animal models outlined in Example 13, animals are treated with signal-sensor polynucleotide for MYC inhibitor D vs. negative control (untranslatable mRNA for MYC inhibitor D) vs. vehicle. For the KRas model addition of the existing transduced OmoMyc model may also be utilized. Animals are then evaluated for gene expression, tumor status or for any of the hallmarks associated with cancer phenotypes or genotypes.

Deliver one or multiple mRNA therapeutics resulting in a protein (native or non-native, intracellular or extracellular) that confers a cytoprotective advantage to normal cells in the setting of cancer therapeutics (both mRNA and non-mRNA)

miRNA-binding sites are used in the 3′UTR of mRNA therapeutics to direct cytotoxic or cytoprotective mRNA therapeutics to specific cells (normal and/or cancerous).

A strong apoptotic signal (i.e., AIFsh—Apoptosis Inducing Factor short isoform, constitutively active (C.A.) caspase 6 (also known as Rev-caspase-6)—is a component of HSV1-tk—herpes simplex virus 1-thymidine kinase) is encoded as the oncology-related polypeptide or “signal” and is encoded along with a series of 3′UTR miR binding sites, such as that for mir-122a, that would make the signal-sensor polynucleotide relatively much more stable in cancerous cells than in normal cells.

Experiments comparing cancer vs. normal hepatic cell lines where the cancer cell lines have a specific miR signature are performed in vitro. SNU449 or HEP3B (human derived HCC cell lines) are used because both have been shown to have “undetectable miR-122a”, whereas normal hepatocytes should have very high miR-122a levels.

A. AIFsh Encoded Polypeptide Study

First a cancer cell is selected which is sensitive to AIFsh signal-sensor polynucleotide (i.e., it results in apoptosis).

Three miR-122a binding sites are encoded into the 3′UTR of an mRNA sequence for AIFsh and the study arms include 2 cell lines (normal hepatocyte, SNU449 or HEP3B)×5 treatments (vehicle alone, signal-sensor polynucleotide untranslatable, signal-sensor polynucleotide AIFsh (no miR BS in 3′UTR), 3′UTR[miR122a BS×3]-signal-sensor polynucleotide untranslatable, 3′UTR[miR122a BS×3]-signal-sensor polynucleotide AIFsh).

The expected result would be significant apoptosis in the face of signal-sensor polynucleotide AIFsh in both normal and cancer (HEP3B or SNU449) cell lines in the absence of any 3′UTR-miR122a BS. However, a significant difference in the relative apoptosis of normal vs. cancer cell lines in the face of 3′UTR [miR122a BS×3]-signal-sensor polynucleotide AIFsh.

Reversibility of the effect is shown with the co-administration of miR122a to the cancer cell line (e.g., through some transduction of the miR122a activity back into the cancer cell line).

B. C.A. Caspase 6 Encoded Polypeptide Study

First a cancer cell is selected which is sensitive to C.A. caspase 6 signal-sensor polynucleotide (i.e., it results in apoptosis).

Three miR-122a binding sites are encoded into the 3′UTR of an mRNA sequence for C.A. caspase 6 and the study arms include 2 cell lines (normal hepatocyte, SNU449 or HEP3B)×5 treatments (vehicle alone, signal-sensor polynucleotide untranslatable, signal-sensor polynucleotide C.A. caspase 6 (no miR BS in 3′UTR), 3′UTR[miR122a BS×3]-signal-sensor polynucleotide untranslatable, 3′UTR[miR122a BS×3]-signal-sensor polynucleotide C.A. caspase 6).

The expected result would be significant apoptosis in the face of signal-sensor polynucleotide C.A. caspase 6 in both normal and cancer (HEP3B or SNU449) cell lines in the absence of any 3′UTR-miR122a BS. However, a significant difference in the relative apoptosis of normal vs. cancer cell lines in the face of 3′UTR [miR122a BS×3]-signal-sensor polynucleotide C.A. caspase 6.

C. HSV1-Tk Encoded Polypeptide Study

First a cancer cell is selected which is sensitive to HSV1-tk signal-sensor polynucleotide (i.e., it results in apoptosis).

Three miR-122a binding sites are encoded into the 3′UTR of an mRNA sequence for HSV1-tk and the study arms include 2 cell lines (normal hepatocyte, SNU449 or HEP3B)×5 treatments (vehicle alone, signal-sensor polynucleotide untranslatable, signal-sensor polynucleotide HSV1-tk (no miR BS in 3′UTR), 3′UTR[miR122a BS×3]-signal-sensor polynucleotide untranslatable, 3′UTR[miR122a BS×3]-signal-sensor polynucleotide HSV1-tk).

The expected result would be significant apoptosis in the face of signal-sensor polynucleotide HSV1-tk in both normal and cancer (HEP3B or SNU449) cell lines in the absence of any 3′UTR-miR122a BS. However, a significant difference in the relative apoptosis of normal vs. cancer cell lines in the face of 3′UTR [miR122a BS×3]-signal-sensor polynucleotide HSV1-tk.

Reversibility of the effect is shown with the co-administration of miR122a to the cancer cell line (e.g., through some transduction of the miR122a activity back into the cancer cell line).

D. In Vivo Study of Signal-Sensor Polynucleotides

In vivo animal studies are performed for AIFsh, C.A. caspase 6 and HSV1-tk using any of the models disclosed herein or a commercially available orthotopic HCC model.

Human embryonic kidney epithelial cells (HEK293A) and primary human hepatocytes (Hepatocytes) were seeded at a density of 200,000 per well in 500 ul cell culture medium (InVitro GRO medium from Celsis, Chicago, Ill.). G-CSF mRNA having an alpha-globin 3′UTR (G-CSF alpha) (mRNA sequence is shown in SEQ ID NO: 6599; polyA tail of approximately 160 nucleotides not shown in sequence; 5′Cap, Cap1; fully modified with 5-methylcytidine and pseudouridine) G-CSF mRNA having an alpha-globin 3′UTR and a miR-122 binding site (G-CSF miR-122) (mRNA sequence is shown in SEQ ID NO: 6600; polyA tail of approximately 160 nucleotides not shown in sequence; 5′Cap, Cap1; fully modified with 5-methylcytidine and pseudouridine) or G-CSF mRNA having an alpha-globin 3′UTR with four miR-122 binding sites with the seed deleted (G-CSF no seed) (mRNA sequence is shown in SEQ ID NO: 6601; polyA tail of approximately 160 nucleotides not shown in sequence; 5′Cap, Cap1; fully modified with 5-methylcytidine and pseudouridine) was tested at a concentration of 250 ng per well in 24 well plates. The expression of G-CSF was measured by ELISA and the results are shown in Table 14.

TABLE 14
miR-122 Binding Sites
HEK293AHepatocytes
ProteinProtein
ExpressionExpression
(ng/mL)(ng/mL)
G-CSF alpha99.858.18
G-CSF miR-12287.670
G-CSF no seed200.28.05

Since HEK293 cells do not express miR-122 there was no down-regulation of G-CSF protein from the sequence containing miR-122. Whereas, the human hepatocytes express high levels of miR-122 and there was a drastic down-regulation of G-CSF protein observed when the G-CSF sequence contained the miR-122 target sequence. Consequently, the mRNA functioned as an auxotrophic mRNA.

Cell lines of liver and lung cancer, such as those described herein, are transfected with MYC inhibitor D modified mRNA in saline or formulated as described herein or in International Application No PCT/US2012/69610, herein incorporated by reference in its entirety. To evaluate the selectivity and/or the effects of therapy with MYC inhibitor D modified mRNA, normal hepatocytes are also transfected with the MYC inhibitor D modified mRNA.

Signal-sensor polynucleotides are formulated in lipid nanoparticles as described herein, known in the art, and/or as described in International Application No PCT/US2012/69610, herein incorporated by reference in its entirety. For tumor delivery, the lipid nanoparticle formulations are adapted for efficient delivery prior to in vitro or in vivo administration. For targeted delivery and/or to reduce toxicity the signal-senor polynucleotides include at least one miR binding site.

The lipid nanoparticle formulations are administered by methods known in the art or described herein (e.g., intravenous, intramuscular and/or intranasal) to liver and lung cancer models (e.g., those described herein and subcutaneous human xenografts in mice, orthotopic human xenografts in mice and transgenic/genetically engineered mouse models).

Signal-sensor polynucleotides are formulated for in vivo delivery in a lung and/or liver cancer model (e.g., those described herein). The signal-sensor polynucleotides are formulated in lipid nanoparticles as described herein, known in the art and/or described in International Application No PCT/US2012/69610, herein incorporated by reference in its entirety.

The lung and/or liver cancer models are analyzed for protein expression, apoptosis, toxicity, efficacy through tumor volume, liver enzyme levels and effect on tumor tissue to evaluate the effect of administration of the formulated signal-sensor polynucleotides on the lung and/or liver cancer models. Assays are used to evaluate protein expression of the signal-sensor polynucleotides. Apoptosis, toxicity, efficacy through tumor volume, liver enzyme levels and tumor tissue are evaluate using common methods known in the art.

Nanoparticle formulations of 98N12-5 (NPA-005) and DLin-KC2-DMA (NPA-003) were tested at varying concentrations to determine the MFI of FL4 or mCherry (mRNA sequence shown in SEQ ID NO: 6602; polyA tail of approximately 160 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and pseudouridine) over a range of doses. The formulations tested are outlined in Table 15. To determine the optimal concentration of nanoparticle formulations of 98N12-5, varying concentrations of formulated modified RNA (100 ng, 10 ng, 1.0 ng, 0.1 ng and 0.01 ng per well) were tested in a 24-well plate of HEK293.

Human embryonic kidney epithelial (HEK293) (LGC standards GmbH, Wesel, Germany) were seeded on 96-well plates (Greiner Bio-one GmbH, Frickenhausen, Germany) and plates were precoated with collagen type1. HEK293 were seeded at a density of 30,000 cells per well in 100 μl cell culture medium. The cell culture medium was DMEM, 10% FCS, adding 2 mM L-Glutamine, 1 mM Sodiumpyruvate and 1× non-essential amino acids (Biochrom AG, Berlin, Germany) and 1.2 mg/ml Sodiumbicarbonate (Sigma-Aldrich, Munich, Germany). Formulations containing mCherry mRNA were added in quadruplicates directly after seeding the cells and incubated.

Cells were harvested by transferring the culture media supernatants to a 96-well Pro-Bind U-bottom plate (Beckton Dickinson GmbH, Heidelberg, Germany). Cells were trypsinized with ½ volume Trypsin/EDTA (Biochrom AG, Berlin, Germany), pooled with respective supernatants and fixed by adding one volume PBS/2% FCS (both Biochrom AG, Berlin, Germany)/0.5% formaldehyde (Merck, Darmstadt, Germany). Samples then were submitted to a flow cytometer measurement with a 532 nm excitation laser and the 610/20 filter for PE-Texas Red in a LSRII cytometer (Beckton Dickinson GmbH, Heidelberg, Germany). The mean fluorescence intensity (MFI) of all events were analyzed and the results of the FL4 MFI of each dose are shown in Table 16. Likewise, to determine the optimal concentration of nanoparticle formulations of DLin-KC2-DMA, varying concentrations of formulated modified RNA (250 ng 100 ng, 10 ng, 1.0 ng, 0.1 ng and 0.01 ng per well) were tested in a 24-well plate of HEK293, and the results of the FL4 MFI of each dose are shown in Table 17. Nanoparticle formulations of DLin-KC2-DMA were also tested at varying concentrations of formulated modified RNA (250 ng, 100 ng and 30 ng per well) in a 24 well plate of HEK293, and the results of the FL4 MFI of each dose are shown in Table 18. A dose of 1 ng/well for 98N12-5 and a dose of 10 ng/well for DLin-KC2-DMA were found to resemble the FL4 MFI of the background.

To determine how close the concentrations resembled the background, we utilized a flow cytometer with optimized filter sets for detection of mCherry expression, and were able to obtain results with increased sensitivity relative to background levels. Doses of 25 ng/well, 0.25 ng/well, 0.025 ng/well and 0.0025 ng/well were analyzed for 98N12-5 (NPA-005) and DLin-KC2-DMA (NPA-003) to determine the MFI of mCherry. As shown in Table 19, the concentration of 0.025 ng/well and lesser concentrations are similar to the background MFI level of mCherry which is about 386.125.

TABLE 15
Formulations
Formulation #NPA-003NPA-005
LipidDLin-KC2-DMA98N12-5
Lipid/RNA2015
wt/wt
Mean size114 nm106 nm
PDI: 0.08PDI: 0.12
TABLE 16
HEK293, NPA-005, 24-well, n = 4
FormulationFL4 MFI
Untreated control0.246
NPA-005 100 ng2.2175
NPA-005 10 ng0.651
NPA-005 1.0 ng0.28425
NPA-005 0.1 ng0.27675
NPA-005 0.01 ng0.2865
TABLE 17
HEK293, NPA-003, 24-well, n = 4
FormulationFL4 MFI
Untreated control0.3225
NPA-003 250 ng2.9575
NPA-003 100 ng1.255
NPA-003 10 ng0.40025
NPA-003 1 ng0.33025
NPA-003 0.1 ng0.34625
NPA-003 0.01 ng0.3475
TABLE 18
HEK293, NPA-003, 24-well, n = 4
FormulationFL4 MFI
Untreated control0.27425
NPA-003 250 ng5.6075
NPA-003 100 ng3.7825
NPA-003 30 ng1.5525
TABLE 19
Concentration and MFI
MFI mCherry
FormulationNPA-003NPA-005
25ng/well11963.2512256.75
0.25ng/well1349.752572.75
0.025ng/well459.50534.75
0.0025ng/well310.75471.75

Formulations of DLin-DMA, DLin-K-DMA, DLin-KC2-DMA, 98N12-5, C12-200 and DLin-MC3-DMA were incubated at a concentration of 60 ng/well or 62.5 ng/well in a plate of HEK293 and 62.5 ng/well in a plate of HepG2 cells for 24 hours to determine the MFI of mCherry (mRNA sequence shown in SEQ ID NO: 6602; polyA tail of approximately 160 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and pseudouridine) for each formulation.

Human embryonic kidney epithelial (HEK293) and hepatocellular carcinoma epithelial (HepG2) cells (LGC standards GmbH, Wesel, Germany) were seeded on 96-well plates (Greiner Bio-one GmbH, Frickenhausen, Germany) and plates for HEK293 cells were precoated with collagen type1. HEK293 were seeded at a density of 30,000 and HepG2 were seeded at a density of 35,000 cells per well in 100 μl cell culture medium. For HEK293 the cell culture medium was DMEM, 10% FCS, adding 2 mM L-Glutamine, 1 mM Sodiumpyruvate and 1× non-essential amino acids (Biochrom AG, Berlin, Germany) and 1.2 mg/ml Sodiumbicarbonate (Sigma-Aldrich, Munich, Germany) and for HepG2 the culture medium was MEM (Gibco Life Technologies, Darmstadt, Germany), 10% FCS adding 2 mM L-Glutamine, 1 mM Sodiumpyruvate and 1× non-essential amino acids (Biochrom AG, Berlin, Germany. Formulations containing mCherry mRNA (mRNA sequence shown in SEQ ID NO: 6602; polyA tail of approximately 160 nucleotides not shown in sequence; 5′ cap, Cap1); were added in quadruplicates directly after seeding the cells and incubated. The mCherry cDNA with the T7 promoter, 5′ untranslated region (UTR) and 3′ UTR used in in vitro transcription (IVT) is given in SEQ ID NO: 6603. The mCherry mRNA was modified with 5meC at each cytidine and pseudouridine replacement at each uridine site.

Cells were harvested by transferring the culture media supernatants to a 96-well Pro-Bind U-bottom plate (Beckton Dickinson GmbH, Heidelberg, Germany). Cells were trypsinized with ½ volume Trypsin/EDTA (Biochrom AG, Berlin, Germany), pooled with respective supernatants and fixed by adding one volume PBS/2% FCS (both Biochrom AG, Berlin, Germany)/0.5% formaldehyde (Merck, Darmstadt, Germany). Samples then were submitted to a flow cytometer measurement with a 532 nm excitation laser and the 610/20 filter for PE-Texas Red in a LSRII cytometer (Beckton Dickinson GmbH, Heidelberg, Germany). The mean fluorescence intensity (MFI) of all events was determined.

The formulations tested are outlined in Table 20 below. As shown in Table 21 for the 60 ng/well and Tables 22, 23, 24 and 25 for the 62.5 ng/well, the formulation of NPA-003 and NPA-018 have the highest mCherry MFI and the formulations of NPA-008, NPA-010 and NPA-013 are most the similar to the background sample mCherry MFI value.

TABLE 20
Formulations
FormulationLipid/RNAMean size
#Lipidwt/wt(nm)
NPA-001DLin-KC2-DMA10155 nm
PDI: 0.08
NPA-002DLin-KC2-DMA15140 nm
PDI: 0.11
NPA-002-2DLin-KC2-DMA15105 nm
PDI: 0.04
NPA-003DLin-KC2-DMA20114 nm
PDI: 0.08
NPA-003-2DLin-KC2-DMA20 95 nm
PDI: 0.02
NPA-00598N12-515127 nm
PDI: 0.12
NPA-00698N12-520126 nm
PDI: 0.08
NPA-007DLin-DMA15148 nm
PDI: 0.09
NPA-008DLin-K-DMA15121 nm
PDI: 0.08
NPA-009C12-20015138 nm
PDI: 0.15
NPA-010DLin-MC3-DMA15126 nm
PDI: 0.09
NPA-012DLin-DMA20 86 nm
PDI: 0.08
NPA-013DLin-K-DMA20104 nm
PDI: 0.03
NPA-014C12-20020101 nm
PDI: 0.06
NPA-015DLin-MC3-DMA20109 nm
PDI: 0.07
TABLE 21
HEK293, 96-well, 60 ng Modified RNA/well
FormulationMFI mCherry
Untreated871.81
NPA-0016407.25
NPA-00214995
NPA-00329499.5
NPA-0053762
NPA-0062676
NPA-0079905.5
NPA-0081648.75
NPA-0092348.25
NPA-0104426.75
NPA-01211466
NPA-0132098.25
NPA-0143194.25
NPA-01514524
TABLE 22
HEK293, 62.5 ng/well
FormulationMFI mCherry
Untreated871.81
NPA-0016407.25
NPA-00214995
NPA-00329499.5
NPA-0053762
NPA-0062676
NPA-0079905.5
NPA-0081648.75
NPA-0092348.25
NPA-0104426.75
NPA-01211466
NPA-0132098.25
NPA-0143194.25
NPA-01514524
TABLE 23
HEK293, 62.5 ng/well
FormulationMFI mCherry
Untreated295
NPA-0073504
NPA-0128286
NPA-0176128
NPA-003-217528
NPA-01834142
NPA-0101095
NPA-0155859
NPA-0193229
TABLE 24
HepG2, 62.5 ng/well
FormulationMFI mCherry
Untreated649.94
NPA-0016006.25
NPA-0028705
NPA-002-215860.25
NPA-00315059.25
NPA-003-228881
NPA-0051676
NPA-0061473
NPA-00715678
NPA-0082976.25
NPA-009961.75
NPA-0103301.75
NPA-01218333.25
NPA-0135853
NPA-0142257
NPA-01516225.75
TABLE 25
HepG2, 62.5 ng/well
FormulationMFI mCherry
Untreated control656
NPA-00716798
NPA-01221993
NPA-01720377
NPA-003-235651
NPA-01840154
NPA-0102496
NPA-01519741
NPA-01916373

mCherry mRNA (SEQ ID NO: 6604; polyA tail of approximately 160 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and pseudouridine) was formulated as a lipid nanoparticle (LNP) using the syringe pump method. The LNP was formulated at a 20:1 weight ratio of total lipid to modified mRNA with a final lipid molar ratio of 50:10:38.5:1.5 (DLin-KC2-DMA: DSPC: Cholesterol: PEG-c-DOMG). The mCherry formulation, listed in Table 26, was characterized by particle size, zeta potential, and encapsulation.

TABLE 26
mCherry Formulation
Formulation #NPA-003-5
Modified mRNAmCherry
Mean size105 nm
PDI: 0.09
Zeta at pH 7.41.8 mV
Encaps.100%
(RiboGr)

The LNP formulation was administered to mice (n=5) intravenously at a modified mRNA dose of 100 ug. Mice were sacrificed at 24 hrs after dosing. The liver and spleen from the mice administered with mCherry modified mRNA formulations were analyzed by immunohistochemistry (IHC), western blot, or fluorescence-activated cell sorting (FACS).

Histology of the liver showed uniform mCherry expression throughout the section, while untreated animals did not express mCherry. Western blots were also used to confirm mCherry expression in the treated animals, whereas mCherry was not detected in the untreated animals. Tubulin was used as a control marker and was detected in both treated and untreated mice, indicating that normal protein expression in hepatocytes was unaffected.

FACS and IHC were also performed on the spleens of mCherry and untreated mice. All leukocyte cell populations were negative for mCherry expression by FACS analysis. By IHC, there were also no observable differences in the spleen in the spleen between mCherry treated and untreated mice.

Experiments are conducted in order to titrate the binding affinity between two cofactors. As used herein, the term “titrate” refers to a method whereby one or more factors are introduced systematically (such as at increasing levels or wherein the one or more factors are systematically modified) to a solution, scenario or series thereof in order to assess a property of interest. In this embodiment, the property of interest is the binding affinity between two cofactors. In one embodiment, constructs encoding the two cofactors are obtained and/or synthesized and a series of mutant constructs are prepared and/or synthesized. Mutant constructs encode cofactor mutants that may include truncated mutants (mutant proteins lacking one or more amino acids from either the N- or C-terminal domains), mutants with regional deletions [proteins wherein internal regions (comprising one or more amino acids) of the protein are absent], mutants with single amino acid substitutions (wherein a normally expressed amino acid is replaced with an alternative amino acid), mutants with one or more additional amino acids added internally or at either terminus, mutants with regional substitutions [proteins wherein internal regions (comprising one or more amino acids) of the protein are substituted with alternative regions (comprising one or more amino acids) and/or combinations of any of these. Mutant constructs are mutated randomly or subjected to targeted mutation based on existing knowledge of the molecular interactions necessary for binding between the two cofactors being investigated.

In some embodiments, a series of mutant proteins are designed such that the mutations follow a progressive pattern along the polypeptide chain. Such series may allow for a better understanding of a particular aspect or feature of the interaction between cofactors. A mutant series may include, for example, the production of a series of mutants, each with a single amino acid substitution, wherein each mutant has a different amino acid along it's polypeptide sequence mutated (e.g. alanine is substituted, thereby eliminating the influence of an amino acid side chain at each position). In another example, a series of mutants are designed such that the mutants in the series comprise truncations of increasing size. In another example, amino acids capable of being post-translationally modified (e.g. phosphorylated, acetylated, ubiquitinated, glycosylated, etc.) in a similar manner may be mutated along the polypeptide sequence in a series of mutants.

For titration experiments with mutant cofactors, a baseline affinity between the two cofactors is established by combining both cofactors under conditions favorable for binding and the binding affinity between the cofactors is assayed. Binding affinity may be assessed using any of a variety of methods known in the art. Such methods may include, but are not limited to Western blot analysis, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), fluorescence resonance energy transfer (FRET), fluorescence recovery after photobleaching (FRAP), fluorescence polarization technologies and/or surface plasmon resonance (SPR) based technologies. For titration, according to one method, a mutant series of one or both cofactors are combined with the two unmutated cofactors (to allow for binding competition between the wild type and mutated proteins). Changes in affinity between the two cofactors in the presence of increasing concentrations of different mutants are assessed and compared and/or plotted against the specific mutations present in the series of mutants that are competing for binding. Alternatively, mutant cofactors in a series are individually combined with a corresponding unmutated binding partner and assessed for binding affinity. Increasing concentrations of the wild type cofactor (corresponding to the mutant cofactor) are introduced and changes in binding between the mutant cofactors and the corresponding unmutated binding partner are assessed. Comparisons are made between the resulting binding curves and the binding curves of other mutants tested.

In some embodiments, titration of the binding affinity between two cofactors is assessed in the presence or absence of increasing concentrations of a third factor. Such a third factor may be an inhibitor or activator of binding between the two cofactors. A series of mutants, as described above, may be generated for a third factor and such a series may be used in titration experiments to assess the effect of mutations on binding between the two cofactors.

Information obtained from titration experiments may be used to design modified mRNA molecules to encode factors that modulate the interaction between cofactors.

In some embodiments, titration experiments are carried out wherein the binding affinity between HIF1 subunits (HIF1-alpha, HIF2-alpha and ARNT) and/or mutated HIF1 subunits and/or other proteins that interact with HIF1 is assessed. Titration experiments may utilize mutant series generated using constructs for one or more of HIF1-alpha, HIF2-alpha, ARNT and/or a third interacting factor. In some embodiments, a mutant series is generated for HIF1-alpha. HIF1-alpha and HIF2-alpha are hydroxylated by HIF hydroxylase enzymes under normal levels of oxygen in the cell, facilitating degradation and/or blocking transcriptional activity. Hydroxylation decreases as oxygen levels drop, allowing HIF1-alpha and/or HIF2-alpha to associate with their cofactor, ARNT leading to elevated expression of genes comprising HIF-response elements (HREs) (Keith, B. et al., HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer. 2011 Dec. 15; 12(1):9-22). In one embodiment, HIF1-alpha mutant series are generated wherein mutations in the series progressively eliminate one or more hydroxylation sites along the polypeptide chain (including, but not limited to proline 402, proline 564 and/or asparagine 803), thereby modulating stability and/or transcriptional activity in mutant versions of HIF1-alpha. In another embodiment, an alternative cofactor, HIF2-alpha is used to generate a mutant series. Such a mutant series may progressively eliminate one or more hydroxylation sites along the polypeptide chain (including, but not limited to proline 405, proline 531 and/or asparagine 847), thereby modulating stability and/or transcriptional activity in mutant versions of HIF2-alpha. In another embodiment, HIF1-alpha and/or HIF2-alpha mutant series are generated that progressively mutate regions necessary for interaction with ARNT, thereby creating mutants with altered abilities to bind ARNT and modulate HIF-dependent gene expression. In another embodiment, ARNT mutant series are generated that progressively mutate regions necessary for interactions with other HIF subunits, thereby creating mutants with altered abilities to bind HIF subunits and modulate HIF-dependent gene expression.

In some embodiments, mutant series are generated for Von Hippel-Landau tumor suppressor protein (pVHL). This protein binds hydroxylated HIF1-alpha and HIF2-alpha, facilitating their ubiquitination and degradation. In one embodiment, mutant series are generated that progressively mutate regions necessary for interaction with HIF1 subunits, thereby creating mutants with altered abilities to bind HIF1 subunits and modulate HIF-dependent gene expression.

Shown in Table 27 and 28 are the transcript sequences and polypeptide sequences (respectively) for protein targets for use in titration experiments. The name and description of the gene encoding the polypeptide of interest are accompanied by the ENSEMBL Transcript ID (ENST) and transcript sequence (Table 27) or the ENSEMBL Protein ID (ENSP) and peptide sequence (Table 28). In some embodiments of the present invention, modified mRNAs may be designed to encode factors that modulate the affinity between HIF subunits and/or HIF-dependent gene expression. Such modified mRNAs may be designed using knowledge gained from titration experiments.

TABLE 27
Transcript sequences for additional targets
for titration experiments
SEQ
TargetENSTID
TargetDescriptionIDTranscript SequenceNO
HIF2-alphahypoxia263734GCTTTACACTCGCGAGCGGACCGCCACACGG6605
inducibleGTCCGGTGCCCGCTGCGCTTCCGCCCCAGCGC
factor 2,TCCTGAGGCGGCCGTACAATCCTCGGCAGTGT
alphaCCTGAGACTGTATGGTCAGCTCAGCCCGGCCT
subunit;CCGACTCCTTCCGACTCCCAGCATTCGAGCCA
endothelialCTTTTTTTTTTCTTTGAAAACTCAGAAAAGTG
PASACTCCTTTTCCAGGGAAAAAGGAACTTGGGTT
domainCCCTTCTCTCCGTCCTCTTTTCGGGTCTGACAG
protein 1CCTCCACCCACTCCTTCCCCGGACCCCGCCTC
CGCGCGCAGGTTCCTCCCAGTCACCTTTCTCC
ACCCCCGCCCCCGCACCTAGCCCGCCGCGCG
CCACCTTCCACCTGACTGCGCGGGGCGCTCGG
GACCTGCGCGCACCTCGGACCTTCACCACCCG
CCCGGGCCGCGGGGAGCGGACGAGGGCCACA
GCCCCCCACCCGCCAGGGAGCCCAGGTGCTC
GGCGTCTGAACGTCTCAAAGGGCCACAGCGA
CAATGACAGCTGACAAGGAGAAGAAAAGGA
GTAGCTCGGAGAGGAGGAAGGAGAAGTCCCG
GGATGCTGCGCGGTGCCGGCGGAGCAAGGAG
ACGGAGGTGTTCTATGAGCTGGCCCATGAGC
TGCCTCTGCCCCACAGTGTGAGCTCCCATCTG
GACAAGGCCTCCATCATGCGACTGGCAATCA
GCTTCCTGCGAACACACAAGCTCCTCTCCTCA
GTTTGCTCTGAAAACGAGTCCGAAGCCGAAG
CTGACCAGCAGATGGACAACTTGTACCTGAA
AGCCTTGGAGGGTTTCATTGCCGTGGTGACCC
AAGATGGCGACATGATCTTTCTGTCAGAAAA
CATCAGCAAGTTCATGGGACTTACACAGGTG
GAGCTAACAGGACATAGTATCTTTGACTTCAC
TCATCCCTGCGACCATGAGGAGATTCGTGAG
AACCTGAGTCTCAAAAATGGCTCTGGTTTTGG
GAAAAAAAGCAAAGACATGTCCACAGAGCG
GGACTTCTTCATGAGGATGAAGTGCACGGTC
ACCAACAGAGGCCGTACTGTCAACCTCAAGT
CAGCCACCTGGAAGGTCTTGCACTGCACGGG
CCAGGTGAAAGTCTACAACAACTGCCCTCCTC
ACAATAGTCTGTGTGGCTACAAGGAGCCCCT
GCTGTCCTGCCTCATCATCATGTGTGAACCAA
TCCAGCACCCATCCCACATGGACATCCCCCTG
GATAGCAAGACCTTCCTGAGCCGCCACAGCA
TGGACATGAAGTTCACCTACTGTGATGACAG
AATCACAGAACTGATTGGTTACCACCCTGAG
GAGCTGCTTGGCCGCTCAGCCTATGAATTCTA
CCATGCGCTAGACTCCGAGAACATGACCAAG
AGTCACCAGAACTTGTGCACCAAGGGTCAGG
TAGTAAGTGGCCAGTACCGGATGCTCGCAAA
GCATGGGGGCTACGTGTGGCTGGAGACCCAG
GGGACGGTCATCTACAACCCTCGCAACCTGC
AGCCCCAGTGCATCATGTGTGTCAACTACGTC
CTGAGTGAGATTGAGAAGAATGACGTGGTGT
TCTCCATGGACCAGACTGAATCCCTGTTCAAG
CCCCACCTGATGGCCATGAACAGCATCTTTGA
TAGCAGTGGCAAGGGGGCTGTGTCTGAGAAG
AGTAACTTCCTATTCACCAAGCTAAAGGAGG
AGCCCGAGGAGCTGGCCCAGCTGGCTCCCAC
CCCAGGAGACGCCATCATCTCTCTGGATTTCG
GGAATCAGAACTTCGAGGAGTCCTCAGCCTA
TGGCAAGGCCATCCTGCCCCCGAGCCAGCCA
TGGGCCACGGAGTTGAGGAGCCACAGCACCC
AGAGCGAGGCTGGGAGCCTGCCTGCCTTCAC
CGTGCCCCAGGCAGCTGCCCCGGGCAGCACC
ACCCCCAGTGCCACCAGCAGCAGCAGCAGCT
GCTCCACGCCCAATAGCCCTGAAGACTATTAC
ACATCTTTGGATAACGACCTGAAGATTGAAG
TGATTGAGAAGCTCTTCGCCATGGACACAGA
GGCCAAGGACCAATGCAGTACCCAGACGGAT
TTCAATGAGCTGGACTTGGAGACACTGGCAC
CCTATATCCCCATGGACGGGGAAGACTTCCA
GCTAAGCCCCATCTGCCCCGAGGAGCGGCTC
TTGGCGGAGAACCCACAGTCCACCCCCCAGC
ACTGCTTCAGTGCCATGACAAACATCTTCCAG
CCACTGGCCCCTGTAGCCCCGCACAGTCCCTT
CCTCCTGGACAAGTTTCAGCAGCAGCTGGAG
AGCAAGAAGACAGAGCCCGAGCACCGGCCCA
TGTCCTCCATCTTCTTTGATGCCGGAAGCAAA
GCATCCCTGCCACCGTGCTGTGGCCAGGCCA
GCACCCCTCTCTCTTCCATGGGGGGCAGATCC
AATACCCAGTGGCCCCCAGATCCACCATTAC
ATTTTGGGCCCACAAAGTGGGCCGTCGGGGA
TCAGCGCACAGAGTTCTTGGGAGCAGCGCCG
TTGGGGCCCCCTGTCTCTCCACCCCATGTCTC
CACCTTCAAGACAAGGTCTGCAAAGGGTTTT
GGGGCTCGAGGCCCAGACGTGCTGAGTCCGG
CCATGGTAGCCCTCTCCAACAAGCTGAAGCT
GAAGCGACAGCTGGAGTATGAAGAGCAAGCC
TTCCAGGACCTGAGCGGGGGGGACCCACCTG
GTGGCAGCACCTCACATTTGATGTGGAAACG
GATGAAGAACCTCAGGGGTGGGAGCTGCCCT
TTGATGCCGGACAAGCCACTGAGCGCAAATG
TACCCAATGATAAGTTCACCCAAAACCCCAT
GAGGGGCCTGGGCCATCCCCTGAGACATCTG
CCGCTGCCACAGCCTCCATCTGCCATCAGTCC
CGGGGAGAACAGCAAGAGCAGGTTCCCCCCA
CAGTGCTACGCCACCCAGTACCAGGACTACA
GCCTGTCGTCAGCCCACAAGGTGTCAGGCAT
GGCAAGCCGGCTGCTCGGGCCCTCATTTGAGT
CCTACCTGCTGCCCGAACTGACCAGATATGAC
TGTGAGGTGAACGTGCCCGTGCTGGGAAGCT
CCACGCTCCTGCAAGGAGGGGACCTCCTCAG
AGCCCTGGACCAGGCCACCTGAGCCAGGCCT
TCTACCTGGGCAGCACCTCTGCCGACGCCGTC
CCACCAGCTTCACTCTCTCCGTCTGTTTTTGCA
ACTAGGTATTTCTAACGCCAGCACACTATTTA
CAAGATGGACTTACCTGGCAGACTTGCCCAG
GTCACCAAGCAGTGGCCTTTTTCTGAGATGCT
CACTTTATTATCCCTATTTTTAAAGTACACAA
TTGTTTTACCTGTTCTGAAATGTTCTTAAATTT
TGTAGGATTTTTTTCCTCCCCACCTTCAATGA
CTTCTAATTTATATTATCCATAGGTTTCTCTCC
CTCCTTCTCCTTCTCACACACAACTGTCCATA
CTAACAAGTTTGGTGCATGTCTGTTCTTCTGT
AGGGAGAAGCTTTAGCTTCATTTTACTAAAAA
GATTCCTCGTTATTGTTGTTGCCAAAGAGAAA
CAAAAATGATTTTGCTTTCCAAGCTTGGTTTG
TGGCGTCTCCCTCGCAGAGCCCTTCTCGTTTC
TTTTTTAAACTAATCACCATATTGTAAATTTC
AGGGTTTTTTTTTTTTTGTTTAAGCTGACTCTT
TGCTCTAATTTTGGAAAAAAAGAAATGTGAA
GGGTCAACTCCAACGTATGTGGTTATCTGTGA
AAGTTGCACAGCGTGGCTTTTCCTAAACTGGT
GTTTTTCCCCCGCATTTGGTGGATTTTTTATTA
TTATTCAAAAACATAACTGAGTTTTTTAAAAG
AGGAGAAAATTTATATCTGGGTTAAGTGTTTA
TCATATATATGGGTACTTTGTAATATCTAAAA
ACTTAGAAACGGAAATGGAATCCTGCTCACA
AAATCACTTTAAGATCTTTTCGAAGCTGTTAA
TTTTTCTTAGTGTTGTGGACACTGCAGACTTG
TCCAGTGCTCCCACGGCCTGTACGGACACTGT
GGAAGGCCTCCCTCTGTCGGCTTTTTGCCATC
TGTGATATGCCATAGGTGTGACAATCCGAGC
AGTGGAGTCATTCAGCGGGAGCACTGCGCGC
TATCCCCTCACATTCTCTATGTACTATGTATGT
ATGTATTATTATTATTGCTGCCAAGAGGGTCT
GATGGCACGTTGTGGGGTCGGGGGGTGGGGC
GGGGAAGTGCTCTAACTTTTCTTAAGGTTTTG
TTGCTAGCCCTTCAAGTGCACTGAGCTATGTG
ACTCGGATGGTCTTTCACACGGCACATTTGGA
CATTTCCAGAACTACCATGAGATGGTTTAGAC
GGGAATTCATGCAAATGAGGGGTCAAAAATG
GTATAGTGACCCCGTCCACGTCCTCCAAGCTC
ACGACCTTGGAGCCCCGTGGAGCTGGACTGA
GGAGGAGGCTGCACAGCGGGAGAGCAGCTG
GTCCAGACCAGCCCTGCAGCCCCCACTCAGC
CGGCAGCCAGATGGCCCCGCAAGGCCTCCAG
GGATGGCCCCTAGCCACAGGCCCTGGCTGAG
GTCTCTGGGTCGGTCAGTGACATGTAGGTAG
GAAGCACTGAAAATAGTGTTCCCAGAGCACT
TTGCAACTCCCTGGGTAAGAGGGACGACACC
TCTGGTTTTTCAATACCAATTACATGGAACTT
TTCTGTAATGGGTACAATGAAGAAGTTTCTAA
AAACACACACAAAGCACATTGGGCCAACTAT
TTAGTAAGCCCGGATAGACTTATTGCCAAAA
ACAAAAAATAGCTTTCAAAAGAAATTTAAGT
TCTATGAGAAATTCCTTAGTCATGGTGTTGCG
TAAATCATATTTTAGCTGCACGGCATTACCCC
ACACAGGGTGGCAGAACTTGAAGGGTTACTG
ACGTGTAAATGCTGGTATTTGATTTCCTGTGT
GTGTTGCCCTGGCATTAAGGGCATTTTACCCT
TGCAGTTTTACTAAAACACTGAAAAATATTCC
AAGCTTCATATTAACCCTACCTGTCAACGTAA
CGATTTCATGAACGTTATTATATTGTCGAATT
CCTACTGACAACATTATAACTGTATGGGAGCT
TAACTTTATAAGGAAATGTATTTTGACACTGG
TATCTTATTAAAGTATTCTGATCCTA
pVHLvon Hippel-256474TGAGTGTTTATGTTTGTAGTTTTAATTGCTCTG6606
LindauAAGTAAATATCTGATTTTCCAATTTCCACCAG
tumorAGTGCTCTGCACATAGTAGGTCTAATTATTTT
suppressorTCCCTCTTTACTAATCACCCATGCCTTGTAAG
AATTCAGTTAGTTGACTTTTTGTACTTTATAA
GCGTGATGATTGGGTGTTCCCGTGTGAGATGC
GCCACCCTCGAACCTTGTTACGACGTCGGCAC
ATTGCGCGTCTGACATGAAGAAAAAAAAAAT
TCAGTTAGTCCACCAGGCACAGTGGCTAAGG
CCTGTAATCCCTGCACTTTGAGAGGCCAAGGC
AGGAGGATCACTTGAACCCAGGAGTTCGAGA
CCAGCCTAGGCAACATAGCGAGACTCCGTTT
CAAACAACAAATAAAAATAATTAGTCGGGCA
TGGTGGTGCGCGCCTACAGTACCAACTACTCG
GGAGGCTGAGGCGAGACGATCGCTTGAGCCA
GGGAGGTCAAGGCTGCAGTGAGCCAAGCTCG
CGCCACTGCACTCCAGCCCGGGCGACAGAGT
GAGACCCTGTCTCAAAAAAAAAAAAAACACC
AAACCTTAGAGGGGCGAAAAAAAATTTTATA
GTGGAAATACAGTAACGAGTTGGCCTAGCCT
CGCCTCCGTTACAACGGCCTACGGTGCTGGA
GGATCCTTCTGCGCACGCGCACAGCCTCCGGC
CGGCTATTTCCGCGAGCGCGTTCCATCCTCTA
CCGAGCGCGCGCGAAGACTACGGAGGTCGAC
TCGGGAGCGCGCACGCAGCTCCGCCCCGCGT
CCGACCCGCGGATCCCGCGGCGTCCGGCCCG
GGTGGTCTGGATCGCGGAGGGAATGCCCCGG
AGGGCGGAGAACTGGGACGAGGCCGAGGTA
GGCGCGGAGGAGGCAGGCGTCGAAGAGTAC
GGCCCTGAAGAAGACGGCGGGGAGGAGTCG
GGCGCCGAGGAGTCCGGCCCGGAAGAGTCCG
GCCCGGAGGAACTGGGCGCCGAGGAGGAGAT
GGAGGCCGGGCGGCCGCGGCCCGTGCTGCGC
TCGGTGAACTCGCGCGAGCCCTCCCAGGTCAT
CTTCTGCAATCGCAGTCCGCGCGTCGTGCTGC
CCGTATGGCTCAACTTCGACGGCGAGCCGCA
GCCCTACCCAACGCTGCCGCCTGGCACGGGC
CGCCGCATCCACAGCTACCGAGGTCACCTTTG
GCTCTTCAGAGATGCAGGGACACACGATGGG
CTTCTGGTTAACCAAACTGAATTATTTGTGCC
ATCTCTCAATGTTGACGGACAGCCTATTTTTG
CCAATATCACACTGCCAGTGTATACTCTGAAA
GAGCGATGCCTCCAGGTTGTCCGGAGCCTAG
TCAAGCCTGAGAATTACAGGAGACTGGACAT
CGTCAGGTCGCTCTACGAAGATCTGGAAGAC
CACCCAAATGTGCAGAAAGACCTGGAGCGGC
TGACACAGGAGCGCATTGCACATCAACGGAT
GGGAGATTGAAGATTTCTGTTGAAACTTACAC
TGTTTCATCTCAGCTTTTGATGGTACTGATGA
GTCTTGATCTAGATACAGGACTGGTTCCTTCC
TTAGTTTCAAAGTGTCTCATTCTCAGAGTAAA
ATAGGCACCATTGCTTAAAAGAAAGTTAACT
GACTTCACTAGGCATTGTGATGTTTAGGGGCA
AACATCACAAAATGTAATTTAATGCCTGCCCA
TTAGAGAAGTATTTATCAGGAGAAGGTGGTG
GCATTTTTGCTTCCTAGTAAGTCAGGACAGCT
TGTATGTAAGGAGGTTTGTATAAGTAATTCAG
TGGGAATTGCAGCATATCGTTTAATTTTAAGA
AGGCATTGGCATCTGCTTTTAATGGATGTATA
ATACATCCATTCTACATCCGTAGCGGTTGGTG
ACTTGTCTGCCTCCTGCTTTGGGAAGACTGAG
GCATCCGTGAGGCAGGGACAAGTCTTTCTCCT
CTTTGAGACCCCAGTGCCTGCACATCATGAGC
CTTCAGTCAGGGTTTGTCAGAGGAACAAACC
AGGGGACACTTTGTTAGAAAGTGCTTAGAGG
TTCTGCCTCTATTTTTGTTGGGGGGTGGGAGA
GGGGACCTTAAAATGTGTACAGTGAACAAAT
GTCTTAAAGGGAATCATTTTTGTAGGAAGCAT
TTTTTATAATTTTCTAAGTCGTGCACTTTCTCG
GTCCACTCTTGTTGAAGTGCTGTTTTATTACT
GTTTCTAAACTAGGATTGACATTCTACAGTTG
TGATAATAGCATTTTTGTAACTTGCCATCCGC
ACAGAAAATACGAGAAAATCTGCATGTTTGA
TTATAGTATTAATGGACAAATAAGTTTTTGCT
AAATGTGAGTATTTCTGTTCCTTTTTGTAAAT
ATGTGACATTCCTGATTGATTTGGGTTTTTTTG
TTGTTGTTGTTTTGTTTTGTTTTGTTTTTTTGAG
ATGGAGTCTCACTCTTGTCACCCAGGCTGGAG
TGCAGTGGCGCCATCTCGGCTCACTGCAACCT
CTGCCTCCTGGGTTCACGTAATCCTCCTGAGT
AGCTGGGATTACAGGCGCCTGCCACCACGCT
GGCCAATTTTTGTACTTTTAGTAGAGACAGTG
TTTCGTCATGTTGGCCAGGCTGGTTTCAAACT
CCTGACCTCAGGTGATCCGCCCACCTCAGCCT
CCCAAAATGGTGGGATTACAGGTGTGTGGGC
CACCGTGCCTGGCTGATTCAGCATTTTTTATC
AGGCAGGACCAGGTGGCACTTCCACCTCCAG
CCTCTGGTCCTACCAATGGATTCATGGAGTAG
CCTGGACTGTTTCATAGTTTTCTAAATGTACA
AATTCTTATAGGCTAGACTTAGATTCATTAAC
TCAAATTCAATGCTTCTATCAGACTCAGTTTT
TTGTAACTAATAGATTTTTTTTTCCACTTTTGT
TCTACTCCTTCCCTAATAGCTTTTTAAAAAAA
TCTCCCCAGTAGAGAAACATTTGGAAAAGAC
AGAAAACTAAAAAGGAAGAAAAAAGATCCC
TATTAGATACACTTCTTAAATACAATCACATT
AACATTTTGAGCTATTTCCTTCCAGCCTTTTTA
GGGCAGATTTTGGTTGGTTTTTACATAGTTGA
GATTGTACTGTTCATACAGTTTTATACCCTTTT
TCATTTAACTTTATAACTTAAATATTGCTCTAT
GTTAGTATAAGCTTTTCACAAACATTAGTATA
GTCTCCCTTTTATAATTAATGTTTGTGGGTATT
TCTTGGCATGCATCTTTAATTCCTTATCCTAGC
CTTTGGGCACAATTCCTGTGCTCAAAAATGAG
AGTGACGGCTGGCATGGTGGCTCCCGCCTGT
AATCCCAGTACTTTGGAAAGCCAAGGTAAGA
GGATTGCTTGAGCCCAGAACTTCAAGATGAG
CCTGGGCTCATAGTGAGAACCCATCTATACA
AAAAATTTTTAAAAATTAGCATGGCGGCACA
CATCTGTAATCCTAGCTACTTGGCAGGCTGAG
GTGAGAAGATCATTGGAGTTTAGGAATTGGA
GGCTGCAGTGAGCCATGAGTATGCCACTGCA
CTCCAGCCTGGGGGACAGAGCAAGACCCTGC
CTCAAAAAAAAAAAAAAAAAAAAAATCAGG
CCGGGCATGGTGGCTCACGCCTGTAATCCCA
GCACTTTGGGAGGTCGAGGTGGGCAGATCAC
CTGAGGTCAGGAGTTCGAGACCAGCCTGGCC
AACATGGTAAAACCCCATTTCTACTAAAAAA
TACAAGAAT
pVHLvon Hippel-345392CCCGCGTCCGACCCGCGGATCCCGCGGCGTC6607
LindauCGGCCCGGGTGGTCTGGATCGCGGAGGGAAT
tumorGCCCCGGAGGGCGGAGAACTGGGACGAGGCC
suppressorGAGGTAGGCGCGGAGGAGGCAGGCGTCGAA
GAGTACGGCCCTGAAGAAGACGGCGGGGAG
GAGTCGGGCGCCGAGGAGTCCGGCCCGGAAG
AGTCCGGCCCGGAGGAACTGGGCGCCGAGGA
GGAGATGGAGGCCGGGCGGCCGCGGCCCGTG
CTGCGCTCGGTGAACTCGCGCGAGCCCTCCCA
GGTCATCTTCTGCAATCGCAGTCCGCGCGTCG
TGCTGCCCGTATGGCTCAACTTCGACGGCGAG
CCGCAGCCCTACCCAACGCTGCCGCCTGGCA
CGGGCCGCCGCATCCACAGCTACCGAGTGTA
TACTCTGAAAGAGCGATGCCTCCAGGTTGTCC
GGAGCCTAGTCAAGCCTGAGAATTACAGGAG
ACTGGACATCGTCAGGTCGCTCTACGAAGAT
CTGGAAGACCACCCAAATGTGCAGAAAGACC
TGGAGCGGCTGACACAGGAGCGCATTGCACA
TCAACGGATGGGAGATTGAAGATTTCTGTTG
AAACTTACACTGTTTCATCTCAGCTTTTGATG
GTACTGATGAGTCTTGATCTAGATACAGGACT
GGTTCCTTCCTTAGTTTCAAAGTGTCTCATTCT
CAGAGTAAAATAGGCACCATTGCTTAAAAGA
AAGTTAACTGACTTCACTAGGCATTGTGATGT
TTAGGGGCAAACATCACAAAATGTAATTTAA
TGCCTGCCCATTAGAGAAGTATTTATCAGGAG
AAGGTGGTGGCATTTTTGCTTCCTAGTAAGTC
AGGACAGCTTGTATGTAAGGAGGTTTGTATA
AGTAATTCAGTGGGAATTGCAGCATATCGTTT
AATTTTAAGAAGGCATTGGCATCTGCTTTTAA
TGGATGTATAATACATCCATTCTACATCCGTA
GCGGTTGGTGACTTGTCTGCCTCCTGCTTTGG
GAAGACTGAGGCATCCGTGAGGCAGGGACAA
GTCTTTCTCCTCTTTGAGACCCCAGTGCCTGC
ACATCATGAGCCTTCAGTCAGGGTTTGTCAGA
GGAACAAACCAGGGGACACTTTGTTAGAAAG
TGCTTAGAGGTTCTGCCTCTATTTTTGTTGGG
GGGTGGGAGAGGGGACCTTAAAATGTGTACA
GTGAACAAATGTCTTAAAGGGAATCATTTTTG
TAGGAAGCATTTTTTATAATTTTCTAAGTCGT
GCACTTTCTCGGTCCACTCTTGTTGAAGTGCT
GTTTTATTACTGTTTCTAAACTAGGATTGACA
TTCTACAGTTGTGATAATAGCATTTTTGTAAC
TTGCCATCCGCACAGAAAATACGAGAAAATC
TGCATGTTTGATTATAGTATTAATGGACAAAT
AAGTTTTTGCTAAATGTGAGTATTTCTGTTCC
TTTTTGTAAATATGTGACATTCCTGATTGATTT
GGGTTTTTTTGTTGTTGTTGTTTTGTTTTGTTTT
GTTTTTTTGAGATGGAGTCTCACTCTTGTCAC
CCAGGCTGGAGTGCAGTGGCGCCATCTCGGC
TCACTGCAACCTCTGCCTCCTGGGTTCACGTA
ATCCTCCTGAGTAGCTGGGATTACAGGCGCCT
GCCACCACGCTGGCCAATTTTTGTACTTTTAG
TAGAGACAGTGTTTCGTCATGTTGGCCAGGCT
GGTTTCAAACTCCTGACCTCAGGTGATCCGCC
CACCTCAGCCTCCCAAAATGGTGGGATTACA
GGTGTGTGGGCCACCGTGCCTGGCTGATTCAG
CATTTTTTATCAGGCAGGACCAGGTGGCACTT
CCACCTCCAGCCTCTGGTCCTACCAATGGATT
CATGGAGTAGCCTGGACTGTTTCATAGTTTTC
TAAATGTACAAATTCTTATAGGCTAGACTTAG
ATTCATTAACTCAAATTCAATGCTTCTATCAG
ACTCAGTTTTTTGTAACTAATAGATTTTTTTTT
CCACTTTTGTTCTACTCCTTCCCTAATAGCTTT
TTAAAAAAATCTCCCCAGTAGAGAAACATTT
GGAAAAGACAGAAAACTAAAAAGGAAGAAA
AAAGATCCCTATTAGATACACTTCTTAAATAC
AATCACATTAACATTTTGAGCTATTTCCTTCC
AGCCTTTTTAGGGCAGATTTTGGTTGGTTTTT
ACATAGTTGAGATTGTACTGTTCATACAGTTT
TATACCCTTTTTCATTTAACTTTATAACTTAAA
TATTGCTCTATGTTAGTATAAGCTTTTCACAA
ACATTAGTATAGTCTCCCTTTTATAATTAATG
TTTGTGGGTATTTCTTGGCATGCATCTTTAATT
CCTTATCCTAGCCTTTGGGCACAATTCCTGTG
CTCAAAAATGAGAGTGACGGCTGGCATGGTG
GCTCCCGCCTGTAATCCCAGTACTTTGGAAAG
CCAAGGTAAGAGGATTGCTTGAGCCCAGAAC
TTCAAGATGAGCCTGGGCTCATAGTGAGAAC
CCATCTATACAAAAAATTTTTAAAAATTAGCA
TGGCGGCACACATCTGTAATCCTAGCTACTTG
GCAGGCTGAGGTGAGAAGATCATTGGAGTTT
AGGAATTGGAGGCTGCAGTGAGCCATGAGTA
TGCCACTGCACTCCAGCCTGGGGGACAGAGC
AAGACCCTGCCTCAAAAAAAAAAAAAAAAA
AAAAA
pVHLvon Hippel-450183GGATCCCGCGGCGTCCGGCCCGGGTGGTCTG6608
LindauGATCGCGGAGGGAATGCCCCGGAGGGCGGAG
tumorAACTGGGACGAGGCCGAGGTAGGCGCGGAG
suppressorGAGGCAGGCGTCGAAGAGTACGGCCCTGAAG
AAGACAGCTACCGAGGTCACCTTTGGCTCTTC
AGAGATGCAGGGACACACGATGGGCTTCTGG
TTAACCAAACTGAATTATTTGTGCCATCTCTC
AATGTTGACGGACAGCCTATTTTTGCCAATAT
CACACTGCCAGTGTATACTCTGAAAGAGCGA
TGCCTCCAGGTTGTCCGGAGCCTAGTCAAGCC
TGAGAATTACAGGAGACTGGACATCGTCAGG
TCGCTCTACGAAGATCTGGAAGACCACCCAA
ATGTGCAGAAAGACCTGGAGCGGCTGACACA
GGAGCGCATTGCACATCAACGGATGGGAGAT
TGAAGATTTCTGTTGAAACTTACACTGTTTCA
TCTCAGCTTTTGATGGTACTGATGAGTCTTGA
TCTAGATACAGGACTGGTTCCTTCCTTAGTTT
CAAAGTGTCTCATTCTCAGAGTAAAATAGGC
ACCATTGCTTAAAAGAAAGTTAACTGACTTCA
CTAGGCATTGTGATGTTTAGGGGCAAACATC
ACAAAATGTAATTTAATGCCTGCCCATTAGAG
AAGTATTTATCAGGAGAAGGTGGTGGCATTTT
TGCTTCCTAGTAAGTCAGGACAGCTTGTATGT
AAGGAGGTTTGTATAAGTAATTCAGTGGGAA
TTGCAGCATATCGTTTAATTTTAAGAAGGCAT
TGGCATCTGCTTTTAATGGATGTATAATACAT
CCATTCTACATCCGTAGCGGTTGGTGACTTGT
CTGCCTCCTGCTTTGGGAAGACTGAGGCATCC
GTGAGGCAGGGACAAGTCTTTCTCCTCTTTGA
GACCCCAGTGCCTGCACATCATGAGCCTTCAG
TCAGGGTTTGTCAGAGGAACAAACCAGGGGA
CACTTTGTTAGAAAGTGCTTAGAGGTTCTGCC
TCTATTTTTGTTGGGGGGTGGGAGAGGGGAC
CTTAAAATGTGTACAGTGAACAAATGTCTTAA
AGGGAATCATTTTTGTAGGAAGCATTTTTTAT
AATTTTCTAAGTCGTGCACTTTCTCGGTCCAC
TCTTGTT
HIF1-alphahypoxia557538ATTTGAAAACTTGGCAACCTTGGATTGGATGG6609
inducibleATTCATATTTCTTAGTATAGAAGTTCTTGATA
factor 1,TAACTGAAAAATTAAGTTAAACACTTAATAA
alphaGTGGTGGTTACTCAGCACTTTTAGATGCTGTT
subunitTATAATAGATGACCTTTTCTAACTAATTTACA
(basic helix-GTTTTTTGAAAGATAACTGAGAGGTTGAGGG
loop-helixACGGAGATTTTCTTCAAGCAATTTTTTTTTTCA
transcriptionTTTTAAATGAGCTCCCAATGTCGGAGTTTGGA
factor)AAACAAATTTGTCTTTTTAAAAGAAGGTCTAG
GAAACTCAAAACCTGAAGAATTGGAAGAAAT
CAGAATAGAAAATGGTAGGATAAGTTCTGAA
CGTCGAAAAGAAAAGTCTCGAGATGCAGCCA
GATCTCGGCGAAGTAAAGAATCTGAAGTTTTT
TATGAGCTTGCTCATCAGTTGCCACTTCCACA
TAATGTGAGTTCGCATCTTGATAAGGCCTCTG
TGATGAGGCTTACCATCAGCTATTTGCGTGTG
AGGAAACTTCTGGATGCTGGTGATTTGGATAT
TGAAGATGACATGAAAGCACAGATGAATTGC
TTTTATTTGAAAGCCTTGGATGGTTTTGTTAT
GGTTCTCACAGATGATGGTGACATGATTTACA
TTTCTGATAATGTGAACAAATACATGGGATTA
ACTCAGTTTGAACTAACTGGACACAGTGTGTT
TGATTTTACTCATCCATGTGACCATGAGGAAA
TGAGAGAAATGCTTACACACAGAAATGGCCT
TGTGAAAAAGGGTAAAGAACAAAACACACA
GCGAAGCTTTTTTCTCAGAATGAAGTGTACCC
TAACTAGCCGAGGAAGAACTATGAACATAAA
GTCTGCAACATGGAAGGTATTGCACTGCACA
GGCCACATTCACGTATATGATACCAACAGTA
ACCAACCTCAGTGTGGGTATAAGAAACCACC
TATGACCTGCTTGGTGCTGATTTGTGAACCCA
TTCCTCACCCATCAAATATTGAAATTCCTTTA
GATAGCAAGACTTTCCTCAGTCGACACAGCCT
GGATATGAAATTTTCTTATTGTGATGAAAGAA
TTACCGAATTGATGGGATATGAGCCAGAAGA
ACTTTTAGGCCGCTCAATTTATGAATATTATC
ATGCTTTGGACTCTGATCATCTGACCAAAACT
CATCATGATATGTTTACTAAAGGACAAGTCAC
CACAGGACAGTACAGGATGCTTGCCAAAAGA
GGTGGATATGTCTGGGTTGAAACTCAAGCAA
CTGTCATATATAACACCAAGAATTCTCAACCA
CAGTGCATTGTATGTGTGAATTACGTTGTGAG
TGGTATTATTCAGCACGACTTGATTTTCTCCC
TTCAACAAACAGAATGTGTCCTTAAACCGGTT
GAATCTTCAGATATGAAAATGACTCAGCTATT
CACCAAAGTTGAATCAGAAGATACAAGTAGC
CTCTTTGACAAACTTAAGAAGGAACCTGATG
CTTTAACTTTGCTGGCCCCAGCCGCTGGAGAC
ACAATCATATCTTTAGATTTTGGCAGCAACGA
CACAGAAACTGATGACCAGCAACTTGAGGAA
GTACCATTATATAATGATGTAATGCTCCCCTC
ACCCAACGAAAAATTACAGAATATAAATTTG
GCAATGTCTCCATTACCCACCGCTGAAACGCC
AAAGCCACTTCGAAGTAGTGCTGACCCTGCA
CTCAATCAAGAAGTTGCATTAAAATTAGAAC
CAAATCCAGAGTCACTGGAACTTTCTTTTACC
ATGCCCCAGATTCAGGATCAGACACCTAGTC
CTTCCGATGGAAGCACTAGACAAAGTTCACC
TGAGCCTAATAGTCCCAGTGAATATTGTTTTT
ATGTGGATAGTGATATGGTCAATGAATTCAA
GTTGGAATTGGTAGAAAAACTTTTTGCTGAAG
ACACAGAAGCAAAGAACCCATTTTCTACTCA
GGACACAGATTTAGACTTGGAGATGTTAGCT
CCCTATATCCCAATGGATGATGACTTCCAGTT
ACGTTCCTTCGATCAGTTGTCACCATTAGAAA
GCAGTTCCGCAAGCCCTGAAAGCGCAAGTCC
TCAAAGCACAGTTACAGTATTCCAGCAGACT
CAAATACAAGAACCTACTGCTAATGCCACCA
CTACCACTGCCACCACTGATGAATTAAAAAC
AGTGACAAAAGACCGTATGGAAGACATTAAA
ATATTGATTGCATCTCCATCTCCTACCCACAT
ACATAAAGAAACTACTAGTGCCACATCATCA
CCATATAGAGATACTCAAAGTCGGACAGCCT
CACCAAACAGAGCAGGAAAAGGAGTCATAG
AACAGACAGAAAAATCTCATCCAAGAAGCCC
TAACGTGTTATCTGTCGCTTTGAGTCAAAGAA
CTACAGTTCCTGAGGAAGAACTAAATCCAAA
GATACTAGCTTTGCAGAATGCTCAGAGAAAG
CGAAAAATGGAACATGATGGTTCACTTTTTCA
AGCAGTAGGAATTGGAACATTATTACAGCAG
CCAGACGATCATGCAGCTACTACATCACTTTC
TTGGAAACGTGTAAAAGGATGCAAATCTAGT
GAACAGAATGGAATGGAGCAAAAGACAATTA
TTTTAATACCCTCTGATTTAGCATGTAGACTG
CTGGGGCAATCAATGGATGAAAGTGGATTAC
CACAGCTGACCAGTTATGATTGTGAAGTTAAT
GCTCCTATACAAGGCAGCAGAAACCTACTGC
AGGGTGAAGAATTACTCAGAGCTTTGGATCA
AGTTAACTGAGCTTTTTCTTAATTTCATTCCTT
TTTTTGGACACTGGTGGCTCATTACCTAAAGC
AGTCTATTTATATTTTCTACATCTAATTTTAGA
AGCCTGGCTACAATACTGCACAAACTTGGTTA
GTTCAATTTTGATCCCCTTTCTACTTAATTTAC
ATTAATGCTCTTTTTTAGTATGTTCTTTAATGC
TGGATCACAGACAGCTCATTTTCTCAGTTTTT
TGGTATTTAAACCATTGCATTGCAGTAGCATC
ATTTTAAAAAATGCACCTTTTTATTTATTTATT
TTTGGCTAGGGAGTTTATCCCTTTTTCGAATT
ATTTTTAAGAAGATGCCAATATAATTTTTGTA
AGAAGGCAGTAACCTTTCATCATGATCATAG
GCAGTTGAAAAATTTTTACACCTTTTTTTTCA
CATTTTACATAAATAATAATGCTTTGCCAGCA
GTACGTGGTAGCCACAATTGCACAATATATTT
TCTTAAAAAATACCAGCAGTTACTCATGGAAT
ATATTCTGCGTTTATAAAACTAGTTTTTAAGA
AGAAATTTTTTTTGGCCTATGAAATTGTTAAA
CCTGGAACATGACATTGTTAATCATATAATAA
TGATTCTTAAATGCTGTATGGTTTATTATTTA
AATGGGTAAAGCCATTTACATAATATAGAAA
GATATGCATATATCTAGAAGG
HIF1-alphahypoxia394997GACAGGAGGATCACCCTCTTCGTCGCTTCGGC6610
inducibleCAGTGTGTCGGGCTGGGCCCTGACAAGCCAC
factor 1,CTGAGGAGAGGCTCGGAGCCGGGCCCGGACC
alphaCCGGCGATTGCCGCCCGCTTCTCTCTAGTCTC
subunitACGAGGGGTTTCCCGCCTCGCACCCCCACCTC
(basic helix-TGGACTTGCCTTTCCTTCTCTTCTCCGCGTGTG
loop-helixGAGGGAGCCAGCGCTTAGGCCGGAGCGAGCC
transcriptionTGGGGGCCGCCCGCCGTGAAGACATCGCGGG
factor)GACCGATTCACCATGGAGGGCGCCGGCGGCG
CGAACGACAAGAAAAATAGGATAAGTTCTGA
ACGTCGAAAAGAAAAGTCTCGAGATGCAGCC
AGATCTCGGCGAAGTAAAGAATCTGAAGTTT
TTTATGAGCTTGCTCATCAGTTGCCACTTCCA
CATAATGTGAGTTCGCATCTTGATAAGGCCTC
TGTGATGAGGCTTACCATCAGCTATTTGCGTG
TGAGGAAACTTCTGGATGCTGGTGATTTGGAT
ATTGAAGATGACATGAAAGCACAGATGAATT
GCTTTTATTTGAAAGCCTTGGATGGTTTTGTT
ATGGTTCTCACAGATGATGGTGACATGATTTA
CATTTCTGATAATGTGAACAAATACATGGGAT
TAACTCAGTTTGAACTAACTGGACACAGTGTG
TTTGATTTTACTCATCCATGTGACCATGAGGA
AATGAGAGAAATGCTTACACACAGAAATGGC
CTTGTGAAAAAGGGTAAAGAACAAAACACAC
AGCGAAGCTTTTTTCTCAGAATGAAGTGTACC
CTAACTAGCCGAGGAAGAACTATGAACATAA
AGTCTGCAACATGGAAGGTATTGCACTGCAC
AGGCCACATTCACGTATATGATACCAACAGT
AACCAACCTCAGTGTGGGTATAAGAAACCAC
CTATGACCTGCTTGGTGCTGATTTGTGAACCC
ATTCCTCACCCATCAAATATTGAAATTCCTTT
AGATAGCAAGACTTTCCTCAGTCGACACAGC
CTGGATATGAAATTTTCTTATTGTGATGAAAG
AATTACCGAATTGATGGGATATGAGCCAGAA
GAACTTTTAGGCCGCTCAATTTATGAATATTA
TCATGCTTTGGACTCTGATCATCTGACCAAAA
CTCATCATGATATGTTTACTAAAGGACAAGTC
ACCACAGGACAGTACAGGATGCTTGCCAAAA
GAGGTGGATATGTCTGGGTTGAAACTCAAGC
AACTGTCATATATAACACCAAGAATTCTCAAC
CACAGTGCATTGTATGTGTGAATTACGTTGTG
AGTGGTATTATTCAGCACGACTTGATTTTCTC
CCTTCAACAAACAGAATGTGTCCTTAAACCG
GTTGAATCTTCAGATATGAAAATGACTCAGCT
ATTCACCAAAGTTGAATCAGAAGATACAAGT
AGCCTCTTTGACAAACTTAAGAAGGAACCTG
ATGCTTTAACTTTGCTGGCCCCAGCCGCTGGA
GACACAATCATATCTTTAGATTTTGGCAGCAA
CGACACAGAAACTGATGACCAGCAACTTGAG
GAAGTACCATTATATAATGATGTAATGCTCCC
CTCACCCAACGAAAAATTACAGAATATAAAT
TTGGCAATGTCTCCATTACCCACCGCTGAAAC
GCCAAAGCCACTTCGAAGTAGTGCTGACCCT
GCACTCAATCAAGAAGTTGCATTAAAATTAG
AACCAAATCCAGAGTCACTGGAACTTTCTTTT
ACCATGCCCCAGATTCAGGATCAGACACCTA
GTCCTTCCGATGGAAGCACTAGACAAAGTTC
ACCTGAGCCTAATAGTCCCAGTGAATATTGTT
TTTATGTGGATAGTGATATGGTCAATGAATTC
AAGTTGGAATTGGTAGAAAAACTTTTTGCTGA
AGACACAGAAGCAAAGAACCCATTTTCTACT
CAGGACACAGATTTAGACTTGGAGATGTTAG
CTCCCTATATCCCAATGGATGATGACTTCCAG
TTACGTTCCTTCGATCAGTTGTCACCATTAGA
AAGCAGTTCCGCAAGCCCTGAAAGCGCAAGT
CCTCAAAGCACAGTTACAGTATTCCAGCAGA
CTCAAATACAAGAACCTACTGCTAATGCCAC
CACTACCACTGCCACCACTGATGAATTAAAA
ACAGTGACAAAAGACCGTATGGAAGACATTA
AAATATTGATTGCATCTCCATCTCCTACCCAC
ATACATAAAGAAACTACTAGTGCCACATCAT
CACCATATAGAGATACTCAAAGTCGGACAGC
CTCACCAAACAGAGCAGGAAAAGGAGTCATA
GAACAGACAGAAAAATCTCATCCAAGAAGCC
CTAACGTGTTATCTGTCGCTTTGAGTCAAAGA
ACTACAGTTCCTGAGGAAGAACTAAATCCAA
AGATACTAGCTTTGCAGAATGCTCAGAGAAA
GCGAAAAATGGAACATGATGGTTCACTTTTTC
AAGCAGTAGGAATTGGAACATTATTACAGCA
GCCAGACGATCATGCAGCTACTACATCACTTT
CTTGGAAACGTGTAAAAGGATGCAAATCTAG
TGAACAGAATGGAATGGAGCAAAAGACAATT
ATTTTAATACCCTCTGATTTAGCATGTAGACT
GCTGGGGCAATCAATGGATGAAAGTGGATTA
CCACAGCTGACCAGTTATGATTGTGAAGTTAA
TGCTCCTATACAAGGCAGCAGAAACCTACTG
CAGGGTGAAGAATTACTCAGAGCTTTGGATC
AAGTTAACTGAGCTTTTTCTTAATTTCATTCCT
TTTTTTGGACACTGGTGGCTCATTACCTAAAG
CAGTCTATTTATATTTTCTACATCTAATTTTAG
AAGCCTGGCTACAATACTGCACAAACTTGGTT
AGTTCAATTTTGATCCCCTTTCTACTTAATTTA
CATTAATGCTCTTTTTTAGTATGTTCTTTAATG
CTGGATCACAGACAGCTCATTTTCTCAGTTTT
TTGGTATTTAAACCATTGCATTGCAGTAGCAT
CATTTTAAAAAATGCACCTTTTTATTTATTTAT
TTTTGGCTAGGGAGTTTATCCCTTTTTCGAATT
ATTTTTAAGAAGATGCCAATATAATTTTTGTA
AGAAGGCAGTAACCTTTCATCATGATCATAG
GCAGTTGAAAAATTTTTACACCTTTTTTTTCA
CATTTTACATAAATAATAATGCTTTGCCAGCA
GTACGTGGTAGCCACAATTGCACAATATATTT
TCTTAAAAAATACCAGCAGTTACTCATGGAAT
ATATTCTGCGTTTATAAAACTAGTTTTTAAGA
AGAAATTTTTTTTGGCCTATGAAATTGTTAAA
CCTGGAACATGACATTGTTAATCATATAATAA
TGATTCTTAAATGCTGTATGGTTTATTATTTA
AATGGGTAAAGCCATTTACATAATATAGAAA
GATATGCATATATCTAGAAGGTATGTGGCATT
TATTTGGATAAAATTCTCAATTCAGAGAAATC
ATCTGATGTTTCTATAGTCACTTTGCCAGCTC
AAAAGAAAACAATACCCTATGTAGTTGTGGA
AGTTTATGCTAATATTGTGTAACTGATATTAA
ACCTAAATGTTCTGCCTACCCTGTTGGTATAA
AGATATTTTGAGCAGACTGTAAACAAGAAAA
AAAAAATCATGCATTCTTAGCAAAATTGCCTA
GTATGTTAATTTGCTCAAAATACAATGTTTGA
TTTTATGCACTTTGTCGCTATTAACATCCTTTT
TTTCATGTAGATTTCAATAATTGAGTAATTTT
AGAAGCATTATTTTAGGAATATATAGTTGTCA
CAGTAAATATCTTGTTTTTTCTATGTACATTGT
ACAAATTTTTCATTCCTTTTGCTCTTTGTGGTT
GGATCTAACACTAACTGTATTGTTTTGTTACA
TCAAATAAACATCTTCTGTGGACCAGG
TABLE 28
Peptide sequences for additional targets for titration experiments
SEQ
TargetENSPID
TargetDescriptionIDProtein SequenceNO
HIF2-hypoxia263734MTADKEKKRSSSERRKEKSRDAARCRRSKETE6611
alphainducibleVFYELAHELPLPHSVSSHLDKASIMRLAISFLRT
factor 2,HKLLSSVCSENESEAEADQQMDNLYLKALEGFI
alphaAVVTQDGDMIFLSENISKFMGLTQVELTGHSIF
subunit;DFTHPCDHEEIRENLSLKNGSGFGKKSKDMSTE
endothelialRDFFMRMKCTVTNRGRTVNLKSATWKVLHCT
PASGQVKVYNNCPPHNSLCGYKEPLLSCLIIMCEPIQ
domainHPSHMDIPLDSKTFLSRHSMDMKFTYCDDRITE
protein 1LIGYHPEELLGRSAYEFYHALDSENMTKSHQNL
CTKGQVVSGQYRMLAKHGGYVWLETQGTVIY
NPRNLQPQCIMCVNYVLSEIEKNDVVFSMDQTE
SLFKPHLMAMNSIFDSSGKGAVSEKSNFLFTKL
KEEPEELAQLAPTPGDAIISLDFGNQNFEESSAY
GKAILPPSQPWATELRSHSTQSEAGSLPAFTVPQ
AAAPGSTTPSATSSSSSCSTPNSPEDYYTSLDND
LKIEVIEKLFAMDTEAKDQCSTQTDFNELDLET
LAPYIPMDGEDFQLSPICPEERLLAENPQSTPQH
CFSAMTNIFQPLAPVAPHSPFLLDKFQQQLESKK
TEPEHRPMSSIFFDAGSKASLPPCCGQASTPLSS
MGGRSNTQWPPDPPLHFGPTKWAVGDQRTEFL
GAAPLGPPVSPPHVSTFKTRSAKGFGARGPDVL
SPAMVALSNKLKLKRQLEYEEQAFQDLSGGDP
PGGSTSHLMWKRMKNLRGGSCPLMPDKPLSAN
VPNDKFTQNPMRGLGHPLRHLPLPQPPSAISPGE
NSKSRFPPQCYATQYQDYSLSSAHKVSGMASR
LLGPSFESYLLPELTRYDCEVNVPVLGSSTLLQG
GDLLRALDQAT
pVHLvon Hippel-256474MPRRAENWDEAEVGAEEAGVEEYGPEEDGGE6612
LindauESGAEESGPEESGPEELGAEEEMEAGRPRPVLRS
tumorVNSREPSQVIFCNRSPRVVLPVWLNFDGEPQPY
suppressorPTLPPGTGRRIHSYRGHLWLFRDAGTHDGLLVN
QTELFVPSLNVDGQPIFANITLPVYTLKERCLQV
VRSLVKPENYRRLDIVRSLYEDLEDHPNVQKDL
ERLTQERIAHQRMGD
pVHLvon Hippel-344757MPRRAENWDEAEVGAEEAGVEEYGPEEDGGE6613
LindauESGAEESGPEESGPEELGAEEEMEAGRPRPVLRS
tumorVNSREPSQVIFCNRSPRVVLPVWLNFDGEPQPY
suppressorPTLPPGTGRRIHSYRVYTLKERCLQVVRSLVKP
ENYRRLDIVRSLYEDLEDHPNVQKDLERLTQER
IAHQRMGD
pVHLvon Hippel-395399MPRRAENWDEAEVGAEEAGVEEYGPEEDSYR6614
LindauGHLWLFRDAGTHDGLLVNQTELFVPSLNVDGQ
tumorPIFANITLPVYTLKERCLQVVRSLVKPENYRRLD
suppressorIVRSLYEDLEDHPNVQKDLERLTQERIAHQRMGD
HIF1-hypoxia451696MRLTISYLRVRKLLDAGDLDIEDDMKAQMNCF6615
alphainducibleYLKALDGFVMVLTDDGDMIYISDNVNKYMGL
factor 1,TQFELTGHSVFDFTHPCDHEEMREMLTHRNGL
alphaVKKGKEQNTQRSFFLRMKCTLTSRGRTMNIKS
subunitATWKVLHCTGHIHVYDTNSNQPQCGYKKPPMT
(basic helix-CLVLICEPIPHPSNIEIPLDSKTFLSRHSLDMKFSY
loop-helixCDERITELMGYEPEELLGRSIYEYYHALDSDHL
transcriptionTKTHHDMFTKGQVTTGQYRMLAKRGGYVWV
factor)ETQATVIYNTKNSQPQCIVCVNYVVSGIIQHDLI
FSLQQTECVLKPVESSDMKMTQLFTKVESEDTS
SLFDKLKKEPDALTLLAPAAGDTIISLDFGSNDT
ETDDQQLEEVPLYNDVMLPSPNEKLQNINLAM
SPLPTAETPKPLRSSADPALNQEVALKLEPNPES
LELSFTMPQIQDQTPSPSDGSTRQSSPEPNSPSEY
CFYVDSDMVNEFKLELVEKLFAEDTEAKNPFST
QDTDLDLEMLAPYIPMDDDFQLRSFDQLSPLES
SSASPESASPQSTVTVFQQTQIQEPTANATTTTA
TTDELKTVTKDRMEDIKILIASPSPTHIHKETTSA
TSSPYRDTQSRTASPNRAGKGVIEQTEKSHPRSP
NVLSVALSQRTTVPEEELNPKILALQNAQRKRK
MEHDGSLFQAVGIGTLLQQPDDHAATTSLSWK
RVKGCKSSEQNGMEQKTIILIPSDLACRLLGQS
MDESGLPQLTSYDCEVNAPIQGSRNLLQGEELL
RALDQVN
HIF1-hypoxia378446MEGAGGANDKKNRISSERRKEKSRDAARSRRS6616
alphainducibleKESEVFYELAHQLPLPHNVSSHLDKASVMRLTI
factor 1,SYLRVRKLLDAGDLDIEDDMKAQMNCFYLKAL
alphaDGFVMVLTDDGDMIYISDNVNKYMGLTQFELT
subunitGHSVFDFTHPCDHEEMREMLTHRNGLVKKGKE
(basic helix-QNTQRSFFLRMKCTLTSRGRTMNIKSATWKVL
loop-helixHCTGHIHVYDTNSNQPQCGYKKPPMTCLVLICE
transcriptionPIPHPSNIEIPLDSKTFLSRHSLDMKFSYCDERITE
factor)LMGYEPEELLGRSIYEYYHALDSDHLTKTHHD
MFTKGQVTTGQYRMLAKRGGYVWVETQATVI
YNTKNSQPQCIVCVNYVVSGIIQHDLIFSLQQTE
CVLKPVESSDMKMTQLFTKVESEDTSSLFDKLK
KEPDALTLLAPAAGDTIISLDFGSNDTETDDQQL
EEVPLYNDVMLPSPNEKLQNINLAMSPLPTAET
PKPLRSSADPALNQEVALKLEPNPESLELSFTMP
QIQDQTPSPSDGSTRQSSPEPNSPSEYCFYVDSD
MVNEFKLELVEKLFAEDTEAKNPFSTQDTDLDL
EMLAPYIPMDDDFQLRSFDQLSPLESSSASPESA
SPQSTVTVFQQTQIQEPTANATTTTATTDELKTV
TKDRMEDIKILIASPSPTHIHKETTSATSSPYRDT
QSRTASPNRAGKGVIEQTEKSHPRSPNVLSVAL
SQRTTVPEEELNPKILALQNAQRKRKMEHDGSL
FQAVGIGTLLQQPDDHAATTSLSWKRVKGCKS
SEQNGMEQKTIILIPSDLACRLLGQSMDESGLPQ
LTSYDCEVNAPIQGSRNLLQGEELLRALDQVN

Materials for Examples 27-33

Table 29 describes the modified mRNA sequences described in Examples 27-33.

TABLE 29
SEQ
ID
TargetmRNA Sequence (polyA tail and 5′cap not shown in sequence)NO
Apoptosis-GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCC6617
inducingACCAUGGAAAAAGUCAGACGAGAGGGGGUUAAGGUGAUGCCCAA
factor shortUGCUAUUGUGCAAUCCGUUGGAGUCAGCAGUGGCAAGUUACUUA
(AIFsh)UCAAGCUGAAAGACGGCAGGAAGGUAGAAACUGACCACAUAGUG
GCAGCUGUGGGCCUGGAGCCCAAUGUUGAGUUGGCCAAGACUGG
UGGCCUGGAAAUAGACUCAGAUUUUGGUGGCUUCCGGGUAAAUG
CAGAGCUACAAGCACGCUCUAACAUCUGGGUGGCAGGAGAUGCU
GCAUGCUUCUACGAUAUAAAGUUGGGAAGGAGGCGGGUAGAGCA
CCAUGAUCACGCUGUUGUGAGUGGAAGAUUGGCUGGAGAAAAUA
UGACUGGAGCUGCUAAGCCGUACUGGCAUCAGUCAAUGUUCUGG
AGUGAUUUGGGCCCCGAUGUUGGCUAUGAAGCUAUUGGUCUUGU
GGACAGUAGUUUGCCCACAGUUGGUGUUUUUGCAAAAGCAACUG
CACAAGACAACCCCAAAUCUGCCACAGAGCAGUCAGGAACUGGUA
UCCGAUCAGAGAGUGAGACAGAGUCCGAGGCCUCAGAAAUUACU
AUUCCUCCCAGCACCCCGGCAGUUCCACAGGCUCCCGUCCAGGGG
GAGGACUACGGCAAAGGUGUCAUCUUCUACCUCAGGGACAAAGU
GGUCGUGGGGAUUGUGCUAUGGAACAUCUUUAACCGAAUGCCAA
UAGCAAGGAAGAUCAUUAAGGACGGUGAGCAGCAUGAAGAUCUC
AAUGAAGUAGCCAAACUAUUCAACAUUCAUGAAGACUGAUAAUA
GGCUGGAGCCUCGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCCC
CCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCCGUGGUCUUUG
AAUAAAGUCUGAGUGGGCGGC
Siah E3GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCC6618
ubiquitinACCAUGAGCCGUCAGACUGCUACAGCAUUACCUACCGGUACCUCG
proteinAAGUGUCCACCAUCCCAGAGGGUGCCUGCCCUGACUGGCACAACU
ligase 1GCAUCCAACAAUGACUUGGCGAGUCUUUUUGAGUGUCCAGUCUG
(SIAH1)CUUUGACUAUGUGUUACCGCCCAUUCUUCAAUGUCAGAGUGGCC
AUCUUGUUUGUAGCAACUGUCGCCCAAAGCUCACAUGUUGUCCA
ACUUGCCGGGGCCCUUUGGGAUCCAUUCGCAACUUGGCUAUGGA
GAAAGUGGCUAAUUCAGUACUUUUCCCCUGUAAAUAUGCGUCUU
CUGGAUGUGAAAUAACUCUGCCACACACAGAAAAAGCAGACCAU
GAAGAGCUCUGUGAGUUUAGGCCUUAUUCCUGUCCGUGCCCUGG
UGCUUCCUGUAAAUGGCAAGGCUCUCUGGAUGCUGUAAUGCCCC
AUCUGAUGCAUCAGCAUAAGUCCAUUACAACCCUACAGGGAGAG
GAUAUAGUUUUUCUUGCUACAGACAUUAAUCUUCCUGGUGCUGU
UGACUGGGUGAUGAUGCAGUCCUGUUUUGGCUUUCACUUCAUGU
UAGUCUUAGAGAAACAGGAAAAAUACGAUGGUCACCAGCAGUUC
UUCGCAAUCGUACAGCUGAUAGGAACACGCAAGCAAGCUGAAAA
UUUUGCUUACCGACUUGAGCUAAAUGGUCAUAGGCGACGAUUGA
CUUGGGAAGCGACUCCUCGAUCUAUUCAUGAAGGAAUUGCAACA
GCCAUUAUGAAUAGCGACUGUCUAGUCUUUGACACCAGCAUUGC
ACAGCUUUUUGCAGAAAAUGGCAAUUUAGGCAUCAAUGUAACUA
UUUCCAUGUGUUGAUAAUAGGCUGGAGCCUCGGUGGCCAUGCUU
CUUGCCCCUUGGGCCUCCCCCCAGCCCCUCCUCCCCUUCCUGCACC
CGUACCCCCGUGGUCUUUGAAUAAAGUCUGAGUGGGCGGC
ConstitivelyGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCC6619
activeACCAUGAUUGAGACAGACAGUGGUGUUGAUGAUGACAUGGCGUG
(C.A.UCAUAAAAUACCAGUGGAGGCCGACUUCUUGUAUGCAUACUCCA
caspase 3CAGCACCUGGUUAUUAUUCUUGGCGAAAUUCAAAGGAUGGCUCC
(alsoUGGUUCAUCCAGUCGCUUUGUGCCAUGCUGAAACAGUAUGCCGA
known asCAAGCUUGAAUUUAUGCACAUUCUUACCCGGGUUAACCGAAAGG
reverseUGGCAACAGAAUUUGAGUCCUUUUCCUUUGACGCUACUUUUCAU
caspase 3GCAAAGAAACAGAUUCCAUGUAUUGUUUCCAUGCUCACAAAAGA
(Rev-ACUCUAUUUUUAUCACGAUGAAGUUGAUGGGGGAUCCCCCAUGG
Caspase 3))AGAACACUGAAAACUCAGUGGAUUCAAAAUCCAUUAAAAAUUUG
GAACCAAAGAUCAUACAUGGAAGCGAAUCAAUGGACUCUGGAAU
AUCCCUGGACAACAGUUAUAAAAUGGAUUAUCCUGAGAUGGGUU
UAUGUAUAAUAAUUAAUAAUAAGAAUUUUCAUAAGAGCACUGGA
AUGACAUCUCGGUCUGGUACAGAUGUCGAUGCAGCAAACCUCAG
GGAAACAUUCAGAAACUUGAAAUAUGAAGUCAGGAAUAAAAAUG
AUCUUACACGUGAAGAAAUUGUGGAAUUGAUGCGUGAUGUUUCU
AAAGAAGAUCACAGCAAAAGGAGCAGUUUUGUUUGUGUGCUUCU
GAGCCAUGGUGAAGAAGGAAUAAUUUUUGGAACAAAUGGACCUG
UUGACCUGAAAAAAAUAACAAACUUUUUCAGAGGGGAUCGUUGU
AGAAGUCUAACUGGAAAACCCAAACUUUUCAUUAUUCAGGCCUG
CCGUGGUACAGAACUGGACUGUGGCAUUGAGACAGACUGAUAAU
AGGCUGGAGCCUCGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCC
CCCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCCGUGGUCUUU
GAAUAAAGUCUGAGUGGGCGGC
GranulysinGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCC6620
ACCAUGGCAACUUGGGCCCUGCUGCUUCUUGCAGCCAUGUUGCUC
GGAAAUCCUGGUCUGGUGUUUUCGCGCCUUUCACCGGAGUACUA
CGAUCUCGCUCGCGCACAUCUGCGCGACGAGGAGAAGUCGUGCCC
AUGUCUCGCACAAGAAGGGCCACAGGGUGACCUUUUGACCAAGA
CGCAAGAACUUGGCAGGGACUACCGAACCUGUCUGACCAUCGUGC
AAAAGCUGAAGAAAAUGGUCGAUAAACCUACCCAAAGAAGCGUG
UCCAACGCAGCGACUCGGGUGUGCCGGACUGGCAGAUCCAGAUG
GCGGGAUGUGUGUAGAAACUUCAUGAGAAGGUACCAGAGCCGUG
UUACUCAGGGACUGGUCGCGGGAGAAACUGCCCAACAGAUUUGC
GAAGAUCUGCGACUCUGUAUUCCUUCAACCGGACCCCUUUGAUA
AUAGGCUGGAGCCUCGGUGGCCAUGCUUCUUGCCCCUUGGGCCUC
CCCCCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCCGUGGUCUU
UGAAUAAAGUCUGAGUGGGCGGC
MYCGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGCC6621
inhibitor DACCAUGACCGAAGAAAACGUCAAGAGAAGAACCCAUAAUGUCCU
CGAGCGCCAGCGGCGCAAUGAGCUCAAGCGCAGCUUCUUUGCACU
CAGGGACCAAAUUCCAGAGUUGGAGAACAACGAAAAGGCCCCGA
AGGUGGUGAUCCUUAAGAAGGCGACUGCCUACAUCCUGUCGGUG
CAGGCUGAGACUCAAAAGCUGAUCUCCGAAAUCGAUCUGCUCCG
GAAACAGAACGAACAACUGAAACACAAACUGGAACAGCUGCGGA
AUUCAUGCUGAUAAUAGGCUGGAGCCUCGGUGGCCAUGCUUCUU
GCCCCUUGGGCCUCCCCCCAGCCCCUCCUCCCCUUCCUGCACCCGU
ACCCCCGUGGUCUUUGAAUAAAGUCUGAGUGGGCGGC

CD1 mice (Harlan Laboratories, South Easton, Mass.) were administered intravenously lipolexed apoptosis-inducing factor short (AIFsh) modified mRNA (mRNA sequence shown in Table 29; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU), fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC/1 mpU), 25% of uridine modified with 2-thiouridine and 25% of cytidine modified with 5-methylcytidine (s2U and 5mC), fully modified with pseudouridine (pU) or fully modified with 1-methylpseudouridine (1mpU). The mice were administered a dose of 2 ug of mRNA complexed with 2 ul Lipofectamine 2000 (LifeTechnologies, Grand Island, N.Y.) in 100 ul sterile basal DMEM medium (w/o additives, LifeTechnologies, Grand Island, N.Y.).

After 6 hours, the animals were sacrificed and serum & spleen are taken. Spleens were transferred to 6-well plates and kept on ice in presence of 1 ml PBS. One spleen was cut with a scalpel several times and with a rubber cell scraper splenocytes were squeezed out until the PBS turns turbid due to cell release.

Leaving fibrous components behind, the cells were transferred to a 100 um cell strainer (BD Biosciences, San Jose, Calif.) sitting on a 12-well cell culture plate. By gravity the cells passed through the cell strainer and were collected beneath in the 12-well culture dish. 1 ml of PBS was transferred with the free-floating splenocytes to an Eppendorf tube and spun for 5 min at 2000 rpm. The PBS was discarded and the cell pellet combined with 500 ul fresh PBS. The splenocytes were resuspended by brief vortexing for 5 mins at 2000 rpm. The PBS was discarded and 1 ml BD Pharmlyse was added to the cell pellet. The splenocytes were resuspended by brief vortexing. The cells were incubated at room temperature for 3 minutes and then spun at 200 rpm for 5 minutes. The cells were washed twice with 500 ul PBS and spun as described above. The cells were resuspended with 500 ul of PBS and spun as described.

250 ul of splenocytes were combined with 1× Pharmlyse buffer and vortexed briefly or resuspended with a pipet and then spun for 2 minutes at 2000 rpm.

In one tube, resuspend cell pellet in 500 ul RIPA buffer with protease inhibitor co*cktail for mammalian cells (BostonBioproducts, Ashland, Mass.) and freeze lysate or continue with BCA assay immediately. In a second tube, add 250 ul FACS staining kit fixation solution (4% formaldehyde; R and D Systems, Minneapolis, Minn.) and then incubate for 10 minutes at room temperature. The cells were washed twice with 500 ul PBS and spun as described above. The cell pellet was resuspended in 500 PBS and stored at 4° C.

Protein lysates were loaded on NuPage SDS-PAGE system (chambers and power supply) with 1.5 mm ready-to-use Bis-Tris gels and 4-12% acrylamide gradient with MOPS-buffer as running aid (all Life Technologies, Grand Island, N.Y.). Each lysate sample was prepared to 40 ul final volume. This sample contained 25 ug protein lysate in variable volume, RIPA buffer to make up volume to 26 ul, 4 ul of 10× reducing agent and 10 ul 4×SDS loading buffer (both from Life Technologies, Grand Island, N.Y.). Samples were heated at 95° C. for 5 min and loaded on the gel. Standard settings were chosen by the manufacturer, 200V, 120 mA and max. 25 W. Run time was 60 min, but no longer than running dye reaching the lower end of the gel.

After the run was terminated, the plastic case was cracked and the encased gel transferred to a ready-to-use nitrocellulose membrane kit and power supply (iBLOT; LifeTechnologies, Grand Island, N.Y.). Using default settings, the protein lysate was transferred by high Ampere electricity from the gel to the membrane.

After the transfer, the membranes were incubated in 5% BSA in 1×TBS for 15 minutes then in 5% BSA in 1×TBS+0.1% Tween for another 15 minutes. Primary antibodies (AIFsh rabbit polyclonal antibody; Abcam, Cambridge, Mass.) against AIFsh proteins were applied in 3 ml of 5% BSA in 1×TBS solution at a 1:500 to 1:2000 dilution for 3 hours at room temperature and gentle agitation on an orbital shaker. Membranes are washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The secondary antibody (Goat anti-rabbit HRP conjugate; Abeam, Cambridge, Mass.) was conjugated to horse radish peroxidase and binds to the primary antibody antibodies. The secondary antibody was diluted of 1:1000 to 1:5000 in 5% BSA in 1×TBS and incubated for 3 hrs at RT.

At the end of incubation time, the membranes were washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The membranes were developed in 5 ml Pierce WestPico Chemiluminescent Substrate (Thermo Fisher, Rockford, Ill.) as directed.

As shown in FIGS. 3A and 3B the Western Blot detected protein around the expected size of 60 kd for each of the 2 samples evaluated for each chemistry.

CD1 mice (Harlan Laboratories, South Easton, Mass.) were administered intravenously lipolexed siah E3 ubiquitin protein ligase 1 (SIAH1) modified mRNA (mRNA sequence shown in SEQ ID NO. 6618 (Table 29); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU), fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC/1mpU), 25% of uridine modified with 2-thiouridine and 25% of cytidine modified with 5-methylcytidine (s2U and 5mC), fully modified with pseudouridine (pU) or fully modified with 1-methylpseudouridine (1mpU). The mice were administered a dose of 2 ug of mRNA complexed with 2 ul Lipofectamine 2000 (LifeTechnologies, Grand Island, N.Y.) in 100 ul sterile basal DMEM medium (w/o additives, LifeTechnologies, Grand Island, N.Y.).

After 6 hours, the animals were sacrificed and serum & spleen are taken. Spleens were transferred to 6-well plates and kept on ice in presence of 1 ml PBS. One spleen was cut with a scalpel several times and with a rubber cell scraper splenocytes were squeezed out until the PBS turns turbid due to cell release.

Leaving fibrous components behind, the cells were transferred to a 100 um cell strainer (BD Biosciences, San Jose, Calif.) sitting on a 12-well cell culture plate. By gravity the cells passed through the cell strainer and were collected beneath in the 12-well culture dish. 1 ml of PBS was transferred with the free-floating splenocytes to an Eppendorf tube and spun for 5 min at 2000 rpm. The PBS was discarded and the cell pellet combined with 500 ul fresh PBS. The splenocytes were resuspended by brief vortexing for 5 mins at 2000 rpm. The PBS was discarded and 1 ml BD Pharmlyse was added to the cell pellet. The splenocytes were resuspended by brief vortexing. The cells were incubated at room temperature for 3 minutes and then spun at 200 rpm for 5 minutes. The cells were washed twice with 500 ul PBS and spun as described above. The cells were resuspended with 500 ul of PBS and spun as described.

250 ul of splenocytes were combined with 1× Pharmlyse buffer and vortexed briefly or resuspended with a pipet and then spun for 2 minutes at 2000 rpm.

In one tube, resuspend cell pellet in 500 ul RIPA buffer with protease inhibitor co*cktail for mammalian cells (BostonBioproducts, Ashland, Mass.) and freeze lysate or continue with BCA assay immediately. In a second tube, add 250 ul FACS staining kit fixation solution (4% formaldehyde; R and D Systems, Minneapolis, Minn.) and then incubate for 10 minutes at room temperature. The cells were washed twice with 500 ul PBS and spun as described above. The cell pellet was resuspended in 500 PBS and stored at 4° C.

Protein lysates were loaded on NuPage SDS-PAGE system (chambers and power supply) with 1.5 mm ready-to-use Bis-Tris gels and 4-12% acrylamide gradient with MOPS-buffer as running aid (all Life Technologies, Grand Island, N.Y.). Each lysate sample was prepared to 40 ul final volume. This sample contained 25 ug protein lysate in variable volume, RIPA buffer to make up volume to 26 ul, 4 ul of 10× reducing agent and 10 ul 4×SDS loading buffer (both from Life Technologies, Grand Island, N.Y.). Samples were heated at 95° C. for 5 min and loaded on the gel. Standard settings were chosen by the manufacturer, 200V, 120 mA and max. 25 W. Run time was 60 min, but no longer than running dye reaching the lower end of the gel.

After the run was terminated, the plastic case was cracked and the encased gel transferred to a ready-to-use nitrocellulose membrane kit and power supply (iBLOT; LifeTechnologies, Grand Island, N.Y.). Using default settings, the protein lysate was transferred by high Ampere electricity from the gel to the membrane.

After the transfer, the membranes were incubated in 5% BSA in 1×TBS for 15 minutes then in 5% BSA in 1×TBS+0.1% Tween for another 15 minutes. Primary antibodies (SIAH1 rabbit polyclonal antibody; Abcam, Cambridge, Mass.) against SIAH1 proteins were applied in 3 ml of 5% BSA in 1×TBS solution at a 1:500 to 1:2000 dilution for 3 hours at room temperature and gentle agitation on an orbital shaker. Membranes are washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The secondary antibody (Goat anti-rabbit HRP conjugate; Abcam, Cambridge, Mass.) was conjugated to horse radish peroxidase and binds to the primary antibody antibodies. The secondary antibody was diluted of 1:1000 to 1:5000 in 5% BSA in 1×TBS and incubated for 3 hrs at RT.

At the end of incubation time, the membranes were washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The membranes were developed in 5 ml Pierce WestPico Chemiluminescent Substrate (Thermo Fisher, Rockford, Ill.) as directed.

As shown in FIGS. 4A and 4B the Western Blot detected protein around the expected size of 31 kd for each of the 2 samples evaluated for each chemistry.

CD1 mice (Harlan Laboratories, South Easton, Mass.) were administered intravenously lipolexed constitutively active (C.A.) caspase 3 (also known as Reverse-Caspase 3 or Rev-Caspase 3) modified mRNA (mRNA sequence shown in SEQ ID NO. 6619 (Table 29); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU), fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC/1mpU), 25% of uridine modified with 2-thiouridine and 25% of cytidine modified with 5-methylcytidine (s2U and 5mC), fully modified with pseudouridine (pU) or fully modified with 1-methylpseudouridine (1mpU). The mice were administered a dose of 2 ug of mRNA complexed with 2 ul Lipofectamine 2000 (LifeTechnologies, Grand Island, N.Y.) in 100 ul sterile basal DMEM medium (w/o additives, LifeTechnologies, Grand Island, N.Y.).

After 6 hours, the animals were sacrificed and serum & spleen are taken. Spleens were transferred to 6-well plates and kept on ice in presence of 1 ml PBS. One spleen was cut with a scalpel several times and with a rubber cell scraper splenocytes were squeezed out until the PBS turns turbid due to cell release.

Leaving fibrous components behind, the cells were transferred to a 100 um cell strainer (BD Biosciences, San Jose, Calif.) sitting on a 12-well cell culture plate. By gravity the cells passed through the cell strainer and were collected beneath in the 12-well culture dish. 1 ml of PBS was transferred with the free-floating splenocytes to an Eppendorf tube and spun for 5 min at 2000 rpm. The PBS was discarded and the cell pellet combined with 500 ul fresh PBS. The splenocytes were resuspended by brief vortexing for 5 mins at 2000 rpm. The PBS was discarded and 1 ml BD Pharmlyse was added to the cell pellet. The splenocytes were resuspended by brief vortexing. The cells were incubated at room temperature for 3 minutes and then spun at 200 rpm for 5 minutes. The cells were washed twice with 500 ul PBS and spun as described above. The cells were resuspended with 500 ul of PBS and spun as described.

250 ul of splenocytes were combined with 1× Pharmlyse buffer and vortexed briefly or resuspended with a pipet and then spun for 2 minutes at 2000 rpm.

In one tube, resuspend cell pellet in 500 ul RIPA buffer with protease inhibitor co*cktail for mammalian cells (BostonBioproducts, Ashland, Mass.) and freeze lysate or continue with BCA assay immediately. In a second tube, add 250 ul FACS staining kit fixation solution (4% formaldehyde; R and D Systems, Minneapolis, Minn.) and then incubate for 10 minutes at room temperature. The cells were washed twice with 500 ul PBS and spun as described above. The cell pellet was resuspended in 500 PBS and stored at 4° C.

Protein lysates were loaded on NuPage SDS-PAGE system (chambers and power supply) with 1.5 mm ready-to-use Bis-Tris gels and 4-12% acrylamide gradient with MOPS-buffer as running aid (all Life Technologies, Grand Island, N.Y.). Each lysate sample was prepared to 40 ul final volume. This sample contained 25 ug protein lysate in variable volume, RIPA buffer to make up volume to 26 ul, 4 ul of 10× reducing agent and 10 ul 4×SDS loading buffer (both from Life Technologies, Grand Island, N.Y.). Samples were heated at 95° C. for 5 min and loaded on the gel. Standard settings were chosen by the manufacturer, 200V, 120 mA and max. 25 W. Run time was 60 min, but no longer than running dye reaching the lower end of the gel.

After the run was terminated, the plastic case was cracked and the encased gel transferred to a ready-to-use nitrocellulose membrane kit and power supply (iBLOT; LifeTechnologies, Grand Island, N.Y.). Using default settings, the protein lysate was transferred by high Ampere electricity from the gel to the membrane.

After the transfer, the membranes were incubated in 5% BSA in 1×TBS for 15 minutes then in 5% BSA in 1×TBS+0.1% Tween for another 15 minutes. Primary antibodies (Caspase 3 rabbit polyclonal antibody; Abcam, Cambridge, Mass.) against target proteins were applied in 3 ml of 5% BSA in 1×TBS solution at a 1:500 to 1:2000 dilution for 3 hours at room temperature and gentle agitation on an orbital shaker. Membranes are washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The secondary antibody (Goat anti-rabbit HRP conjugate; Abcam, Cambridge, Mass.) was conjugated to horse radish peroxidase and binds to the primary antibody antibodies. The secondary antibody was diluted of 1:1000 to 1:5000 in 5% BSA in 1×TBS and incubated for 3 hrs at RT.

At the end of incubation time, the membranes were washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The membranes were developed in 5 ml Pierce WestPico Chemiluminescent Substrate (Thermo Fisher, Rockford, Ill.) as directed.

As shown in FIGS. 5A and 5B the Western Blot detected protein around the expected size of 32 kd for each of the 2 samples evaluated for each chemistry.

CD1 mice (Harlan Laboratories, South Easton, Mass.) were administered intravenously lipolexed granulysin mRNA (mRNA sequence shown in SEQ ID NO. 6620 (Table 29); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU), fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC/1mpU), 25% of uridine modified with 2-thiouridine and 25% of cytidine modified with 5-methylcytidine (s2U and 5mC), fully modified with pseudouridine (pU) or fully modified with 1-methylpseudouridine (1mpU). The mice were administered a dose of 2 ug of mRNA complexed with 2 ul Lipofectamine 2000 (LifeTechnologies, Grand Island, N.Y.) in 100 ul sterile basal DMEM medium (w/o additives, LifeTechnologies, Grand Island, N.Y.).

After 6 hours, the animals were sacrificed and serum & spleen are taken. Spleens were transferred to 6-well plates and kept on ice in presence of 1 ml PBS. One spleen was cut with a scalpel several times and with a rubber cell scraper splenocytes were squeezed out until the PBS turns turbid due to cell release.

Leaving fibrous components behind, the cells were transferred to a 100 um cell strainer (BD Biosciences, San Jose, Calif.) sitting on a 12-well cell culture plate. By gravity the cells passed through the cell strainer and were collected beneath in the 12-well culture dish. 1 ml of PBS was transferred with the free-floating splenocytes to an Eppendorf tube and spun for 5 min at 2000 rpm. The PBS was discarded and the cell pellet combined with 500 ul fresh PBS. The splenocytes were resuspended by brief vortexing for 5 mins at 2000 rpm. The PBS was discarded and 1 ml BD Pharmlyse was added to the cell pellet. The splenocytes were resuspended by brief vortexing. The cells were incubated at room temperature for 3 minutes and then spun at 200 rpm for 5 minutes. The cells were washed twice with 500 ul PBS and spun as described above. The cells were resuspended with 500 ul of PBS and spun as described.

250 ul of splenocytes were combined with 1× Pharmlyse buffer and vortexed briefly or resuspended with a pipet and then spun for 2 minutes at 2000 rpm.

In one tube, resuspend cell pellet in 500 ul RIPA buffer with protease inhibitor co*cktail for mammalian cells (BostonBioproducts, Ashland, Mass.) and freeze lysate or continue with BCA assay immediately. In a second tube, add 250 ul FACS staining kit fixation solution (4% formaldehyde; R and D Systems, Minneapolis, Minn.) and then incubate for 10 minutes at room temperature. The cells were washed twice with 500 ul PBS and spun as described above. The cell pellet was resuspended in 500 PBS and stored at 4° C.

Protein lysates were loaded on NuPage SDS-PAGE system (chambers and power supply) with 1.5 mm ready-to-use Bis-Tris gels and 4-12% acrylamide gradient with MOPS-buffer as running aid (all Life Technologies, Grand Island, N.Y.). Each lysate sample was prepared to 40 ul final volume. This sample contained 25 ug protein lysate in variable volume, RIPA buffer to make up volume to 26 ul, 4 ul of 10× reducing agent and 10 ul 4×SDS loading buffer (both from Life Technologies, Grand Island, N.Y.). Samples were heated at 95° C. for 5 min and loaded on the gel. Standard settings were chosen by the manufacturer, 200V, 120 mA and max. 25 W. Run time was 60 min, but no longer than running dye reaching the lower end of the gel.

After the run was terminated, the plastic case was cracked and the encased gel transferred to a ready-to-use nitrocellulose membrane kit and power supply (iBLOT; LifeTechnologies, Grand Island, N.Y.). Using default settings, the protein lysate was transferred by high Ampere electricity from the gel to the membrane.

After the transfer, the membranes were incubated in 5% BSA in 1×TBS for 15 minutes then in 5% BSA in 1×TBS+0.1% Tween for another 15 minutes. Primary antibodies (Granulysin mouse monoclonal antibody; Abcam, Cambridge, Mass.) against granulysin proteins were applied in 3 ml of 5% BSA in 1×TBS solution at a 1:500 to 1:2000 dilution for 3 hours at room temperature and gentle agitation on an orbital shaker. Membranes are washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The secondary antibody (Donkey anti-mouse HRP conjugate; Abcam, Cambridge, Mass.) was conjugated to horse radish peroxidase and binds to the primary antibody antibodies. The secondary antibody was diluted of 1:1000 to 1:5000 in 5% BSA in 1×TBS and incubated for 3 hrs at RT.

At the end of incubation time, the membranes were washed 3 times with 1×TBS/0.1% Tween, 5 minutes each time with gentle agitation. The membranes were developed in 5 ml Pierce WestPico Chemiluminescent Substrate (Thermo Fisher, Rockford, Ill.) as directed.

As shown in FIGS. 6A and 6B the Western Blot detected protein around the expected size of 16 kd for each of the 2 samples evaluated for each chemistry.

Proteins can be evaluated using liquid chromatography-mass spectrometry in tandem with mass spectrometry (LC-MS/MS) with quantitative LC-multiple reaction monitoring (MRM) in order to confirm the identity of the peptide.

The identity of any protein target described herein can be evaluated using the liquid chromatography-mass spectrometry in tandem with mass spectrometry (LC-MS/MS) with quantitative LC-multiple reaction monitoring (MRM) Assay (Biognosys AG, Schlieren Switzerland). HeLa cell lysates containing protein expressed from modified mRNA are evaluated using LC-MS/MS with quantitative LC-MRM Assay (Biognosys, Schlieren Switzerland) in order to confirm the identity of the peptides in the cell lysates. The identified peptide fragments are compared against known proteins including isoforms using methods known and/or described in the art.

A. Sample Preparation

Protein in each sample in lysis buffer is reduced by incubation for 1 hour at 37° C. with 5 mM tris(2-carboxyethyl)phosphine (TCEP). Alkylation is carried out using 10 mM iodoacetamide for 30 minutes in the dark at room temperature. Proteins are digested to peptides using trypsin (sequence grade, Promega Corporation, Madison, Wis.) at a protease: protein ratio of 1:50. Digestion is carried out overnight at 37° C. (total digestion time is 12 hours). Peptides are cleaned up for mass spectrometric analysis using C18 spin columns (The Nest Group, Southborough, Mass.) according to the manufacturer's instructions. Peptides are dried down to complete dryness and resuspended in LC solvent A (1% acetonitrile, 0.1% formic acid (FA)). All solvents are HPLC-grade from SIGMA-ALDRICH® (St. Louis, Mo.) and all chemicals, where not stated otherwise, are obtained from SIGMA-ALDRICH® (St. Louis, Mo.).

B. LC-MS/MS and LC-MRM

Peptides are injected to a packed C18 column (Magic AQ, 3 um particle size, 200 Å pore size, Michrom Bioresources, Inc (Auburn, Calif.); 11 cm column length, 75 um inner diameter, New Objective (Woburn, Mass.)) on a Proxeon Easy nLC nano-liquid chromatography system for all mass spectrometric analysis. LC solvents are A: 1% acetonitrile in water with 0.1% FA; B: 3% water in acetonitrile with 0.1% FA. The LC gradient for shotgun analysis is 5-35% solvent B in 120 minutes followed by 35-100% solvent B in 2 minutes and 100% solvent B for 8 minutes (total gradient length is 130 minutes). LC-MS/MS shotgun runs for peptide discovery are carried out on a Thermo Scientific (Thermo Fisher Scientific) (Billerica, Mass.) Q Exactive mass spectrometer equipped with a standard nano-electrospray source. The LC gradient for LC-MRM is 5-35% solvent B in 30 minutes followed by 35-100% solvent B in 2 minutes and 100% solvent B for 8 minutes (total gradient length is 40 minutes). The Thermo Scientific (Thermo Fisher Scientific) (Billerica, Mass.) TSQ Vantage triple quadrupole mass spectrometer is equipped with a standard nano-electrospray source. In unscheduled MRM mode for recalibration it is operated at a dwell time of 20 ms per transition. For relative quantification of the peptides across samples, the TSQ Vantage is operated in scheduled MRM mode with an acquisition window length of 4 minutes. The LC eluent is electrosprayed at 1.9 kV and MRM analysis is performed using a Q1 peak width of 0.7 Da. Collision energies are calculated for the TSQ Vantage by a linear regression according to the vendor's specifications.

C. Assay Design, Data Processing and Analysis

For the generation of LC-MRM assays, the 12 most intense fragment ions from LC-MS/MS analysis are measured in scheduled LC-MRM mode and data were processed using MQUEST® (Cluetec, Karlsruhe, Germany), the scoring part of mProphet (Reiter et al, mProphet: Automated data processing and statistical validation for large-scale SRM experiments, Nature Methods, 2011 (8), 430-435; the contents of which are herein incorporated by reference). Assays were validated manually, exact fragment intensities are determined and iRTs (indexed retention times) are assigned relative to Biognosys's iRT-peptides (Esther et al. Using iRT, a normalized retention time for more targeted measurement of peptides, Proteomics, 2012 (12), 1111-1121; the contents of which are herein incorporated by reference).

For the relative quantification of the peptides across the sample series the 8 most intense transitions of each assay are measured across the sample series. Data analysis is carried out using SpectroDive™ (Biognosys, Schlieren Switzerland). Total peak areas are compared for the selected peptides and a false discover rate of 0.05 is applied. Peptides with a Qvalue below 0.05 are excluded and considered not detected in the respective sample.

Cell lysates containing protein produced from siah E3 ubiquitin protein ligase 1 (SIAH1) modified mRNA (mRNA sequence shown in SEQ ID NO. 6618 (Table 29); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), MYC inhibitor D (a unique dominant-negative 90 amino acid protein comprised of the human c-Myc) modified mRNA (mRNA sequence shown in SEQ ID NO. 6621 (Table 29); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), fully modified with 5-methylcytidine and pseudouridine (5mC and pU), fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1 mpU), modified where 25% of uridine modified with 2-thiouridine and 25% of cytidine modified with 5-methylcytidine (s2U and 5mC), fully modified with pseudouridine (pU), or fully modified with 1-methylpseudouridine (1mpU) were evaluated using the LC-MS/MS with quantitative LC-MRM as described in Example 31. Peptide fragments identified for the evaluated proteins are shown in Table 30.

TABLE 30
Proteins and Peptide Fragment Sequences
Peptide5mC5mCs2U
Fragmentandandand
SEQ ID NOpU1mpU5mCpU1mpU
SIAH1
GPLGSIR6622YESYESYES
MYC INHIBITOR D
ATAYILSVQAET6623YESYESYESYESYES
QK
KATAYILSVQAE6624YESYESYESYESYES
TQK
LISEIDLLRK6625YESYESYESYESYES

Cell lysates containing protein produced from granulysin mRNA (mRNA sequence shown in Table 29; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) fully modified with 1-methylpseudouridine (1mpU) were evaluated using the LC-MS/MS with quantitative LC-MRM as described in Example 31. Peptide fragments identified for the evaluated proteins are shown in Table 31. In Table 31, “Uniprot ID” refers to the protein identifier from the UniProt database when the peptide fragment sequences were blasted against all review proteins in the database.

TABLE 31
Proteins and Peptide Fragment Sequences
Peptide
Fragment
SEQ ID NOUniprot ID
GRANULYSIN
SCPCLAQEGPQGDLLTK6626P22749

Using the animal models outlined in Example 13, animals are treated with signal-sensor polynucleotide encoding for a dominant negative STAT3 molecule or a dominant negative Akt molecule whose expression has been shown to interfere with PI-3 kinase induced oncogenic transformation, including in glioblastoma cells (Vogt and Hart, Cancer Discov, 2011 1:481-486; herein included by reference in its entirety). Animals are injected with mRNA encoding dominant negative STAT3 mRNA vs dominant negative Akt mRNA vs negative control mRNA (non-translated version of the same mRNA containing multiple stop codons) vs vehicle using an appropriate route of delivery and formulation. Animals are then evaluated for gene expression, tumor status or for any of the hallmarks associated with cancer phenotypes or genotypes. Other examples of dominant negative approaches for cancer are outlined and could similarly be used with modified mRNA (Moss and Lemoine Chapter 15 RNA Interference and Dominant Negative Approaches in Viral Therapy of Cancer Harrington et al., eds. Wiley & Sons; herein incorporated by reference in its entirety).

Using the animal models outlined in Example 13, animals are treated with signal-sensor polynucleotide encoding for a dominant negative hTERT whose expression has been shown to interfere with telomerase activity and lead to apoptosis of cancer cells (Agrawal et al. 2012 Recent Pat Anticancer Drug Discov 7:102-117, Samy et al. 2012 Mol Cancer Ther 11:2384-2393, Nguyen et al. 2009 Cell Cycle. 8:3227-3233; all herein included by reference in their entirety). Telomerase, a specialised RNA-directed DNA polymerase extends and stabilises the telomeres at the ends of the eukaryotic chromosomes. The progressive loss of telomeres results in limited number of cell divisions and has been linked to the mechanism of human cellular ageing. Tumour cells marked by indefinite proliferation have stable telomere length maintained by telomerase. The differential expression of the telomerase enzyme in normal and cancer cells have led to the evolution of tumour specific anti-telomerase approaches which inhibit the telomerase enzyme activity so as to destabilise and shorten the telomeres leading to senescence in cancer cells. One such approach is to use modified mRNA to express a dominant negative hTERT. As such animals are injected with mRNA encoding dominant negative hTERT mRNA vs negative control mRNA (non-translated version of the same mRNA containing multiple stop codons) vs vehicle using an appropriate route of delivery and formulation. Animals are then evaluated for gene expression, tumor status or for any of the hallmarks associated with cancer phenotypes or genotypes. Other examples of dominant negative approaches for cancer are outlined and could similarly be used with modified mRNA (Moss and Lemoine Chapter 15 RNA Interference and Dominant Negative Approaches in Viral Therapy of Cancer Harrington et al., eds. Wiley & Sons; herein incorporated by reference in its entirety).

Using the animal models outlined in Example 13, animals are treated with signal-sensor polynucleotide encoding for a dominant negative survivin (C84A and others) whose expression has been shown to lead to apoptosis of cancer cells (Cheung et al. 2010 Cancer Cell Int. 10:36; herein included by reference in its entirety). Survivin is a member of the inhibitor-of-apoptosis (IAP) family which is widely expressed by many different cancers. Overexpression of survivin is associated with drug resistance in cancer cells, and reduced patient survival after chemotherapy and radiotherapy. Agents that antagonize the function of survivin hold promise for treating many forms of cancer. One such approach is to use modified mRNA to express a dominant negative survivin (C84A mutation is one described example). As such animals are injected with mRNA encoding dominant negative survivin mRNA vs negative control mRNA (non-translated version of the same mRNA containing multiple stop codons) vs vehicle using an appropriate route of delivery and formulation. Animals are then evaluated for gene expression, tumor status or for any of the hallmarks associated with cancer phenotypes or genotypes. Other examples of dominant negative approaches for cancer are outlined and could similarly be used with modified mRNA (Moss and Lemoine Chapter 15_RNA Interference and Dominant Negative Approaches in Viral Therapy of Cancer Harrington et al., eds. Wiley & Sons; herein incorporated by reference in its entirety).

Human embryonic kidney epithelial cells (HEK293A), or antigen presenting cells or cell lines with highly expressed mir-142/146, such as monocyte-derived dendritic cells (MDDC) or PBMC, are seeded at a density of 200,000 per well in 500 ul cell culture medium (InVitro GRO medium from Celsis, Chicago, Ill.). G-CSF mRNA (mRNA sequence is shown in SEQ ID NO: 6595; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) G-CSF mRNA having a miR-142-5p binding site (G-CSF miR-142-5p) (cDNA sequence is shown in SEQ ID NO:6627; mRNA sequence is shown in SEQ ID NO: 6628, polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1), G-CSF mRNA having a seed sequence from miR-142-5p binding site (G-CSF miR-142-5p-seed) (cDNA sequence is shown in SEQ ID NO. 6629; mRNA sequence is shown in SEQ ID NO: 6630; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) G-CSF mRNA having a miR-142-5p binding site without the seed sequence (G-CSF miR-142-5p-seedless) (cDNA sequence is shown in SEQ ID NO: 6631, mRNA sequence is shown in SEQ ID NO: 6632; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) G-CSF mRNA having a miR-142-3p binding site (G-CSF miR-142-3p) (cDNA sequence is shown in SEQ ID NO: 6633, mRNA sequence is shown in SEQ ID NO: 6634; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1; G-CSF mRNA having a seed sequence from miR-142-3p binding site (G-CSF miR-142-3p-seed) (cDNA sequence is shown in SEQ ID NO: 6635, mRNA sequence is shown in SEQ ID NO: 6636; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) G-CSF mRNA having a miR-142-3p binding site without the seed sequence (G-CSF miR-142-3p-seedless) (cDNA sequence is shown in SEQ ID NO: 6637; mRNA sequence is shown in SEQ ID NO: 6638; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) G-CSF mRNA having a miR-146a binding site (G-CSF miR-146a) (cDNA sequence is shown in SEQ ID NO. 6639, mRNA sequence is shown in SEQ ID NO: 6640; polyA tail of at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) G-CSF mRNA having a seed sequence from miR-146a binding site (G-CSF miR-146a-seed) (cDNA sequence is shown in SEQ ID NO. 6641, mRNA sequence is shown in SEQ ID NO:6642; polyA tail at least 140 nucleotides not shown in sequence; 5′Cap, Cap1) or G-CSF mRNA having a miR-146a binding site without the seed sequence (G-CSF miR-146a-seedless) (cDNA sequence is shown in SEQ ID NO. 6643, mRNA sequence is shown in SEQ ID NO: 6644; polyA tail at least nucleotides not shown in sequence; 5′Cap, Cap1) are tested at a concentration of 250 ng per well in 24 well plates. The mRNA sequences are evaluated with various chemical modifications described herein and/or known in the art including, fully modified with 5-methylcytidine and pseudouridine, fully modified with 5-methylcytidine and 1-methylpseudouridine, fully modified with pseudouridine, fully modified with 1-methylpseudouridine and where 25% of the uridine residues are modified with 2-thiouridine and 25% of the cytidine residues are modified with 5-methylcytidine. The expression of G-CSF in each sample is measured by ELISA.

Shown in Table 32 are the DNA and mRNA G-CSF sequences with the miR binding sites described above. In the table, the start codon of each sequence is underlined.

TABLE 32
G-CSF constructs with miR binding sites
SEQ
ID
NO.DescriptionSEQ
6627DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
142-5pCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
CTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTAGTAGTGCTTTCTACTTTATGTGGTCTTTGAATAAAGCCTGA
GTAGGAAGGCGGCCGCTCGAGCATGCATCTAGA
6628mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACCAUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGG
G-CSF miR-CCCUGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAA
142-5pGAAGCGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUU
CCUUUUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAU
GGAGCCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUG
CCAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUC
CCUGGGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUG
GCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCA
GGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCC
CGACGCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACA
ACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCU
GCAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUC
AGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCA
UUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCC
GUGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGC
CCUUCUUCUCUCCCUUGCACCUGUACCUCUAGUAGUGCUUUCUA
CUUUAUGUGGUCUUUGAAUAAAGCCUGAGUAGGAAG
6629DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
142-5p-seedCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
CTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTACTTTATTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCGGC
CGCTCGAGCATGCATCTAGA
6630mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACC
G-CSF miR-AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGGCCCU
142-5p-seedGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAG
CGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUU
UUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAUGGAG
CCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUGCCAU
CCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUCCCUG
GGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUGGCAG
GGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCAGGGA
CUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGAC
GCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACAACCA
UCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCUGCAG
CCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUCAGCG
CAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCAUUUU
UGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCG
UGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCC
CUUCUUCUCUCCCUUGCACCUGUACCUCUACUUUAUUGGUCUUU
GAAUAAAGCCUGAGUAGGAAG
6631DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
142-5p-CCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
seedlessCTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTAGTAGTGCTTTCTGTGGTCTTTGAATAAAGCCTGAGTAGGAA
GGCGGCCGCTCGAGCATGCATCTAGA
6632mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACC
G-CSF miR-AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGGCCCU
142-5p-GCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAG
seedlessCGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUU
UUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAUGGAG
CCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUGCCAU
CCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUCCCUG
GGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUGGCAG
GGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCAGGGA
CUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGAC
GCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACAACCA
UCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCUGCAG
CCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUCAGCG
CAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCAUUUU
UGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCG
UGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCC
CUUCUUCUCUCCCUUGCACCUGUACCUCUAGUAGUGCUUUCUGU
GGUCUUUGAAUAAAGCCUGAGUAGGAAG
6633DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
142-3pCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
CTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTTCCATAAAGTAGGAAACACTACATGGTCTTTGAATAAAGCCT
GAGTAGGAAGGCGGCCGCTCGAGCATGCATCTAGA
6634mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACC
G-CSF miR-AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGGCCCU
142-3pGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAG
CGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUU
UUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAUGGAG
CCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUGCCAU
CCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUCCCUG
GGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUGGCAG
GGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCAGGGA
CUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGAC
GCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACAACCA
UCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCUGCAG
CCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUCAGCG
CAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCAUUUU
UGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCG
UGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCC
CUUCUUCUCUCCCUUGCACCUGUACCUCUUCCAUAAAGUAGGAA
ACACUACAUGGUCUUUGAAUAAAGCCUGAGUAGGAAG
6635DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
142-3p-seedCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
CTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTACACTACTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCGGC
CGCTCGAGCATGCATCTAGA
6636mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACC
G-CSF miR-AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGGCCCU
142-3p-seedGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAG
CGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUU
UUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAUGGAG
CCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUGCCAU
CCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUCCCUG
GGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUGGCAG
GGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCAGGGA
CUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGAC
GCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACAACCA
UCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCUGCAG
CCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUCAGCG
CAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCAUUUU
UGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCG
UGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCC
CUUCUUCUCUCCCUUGCACCUGUACCUCUACACUACUGGUCUUU
GAAUAAAGCCUGAGUAGGAAG
6637DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
142-3p-CCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
seedlessCTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTTCCATAAAGTAGGAAATGGTCTTTGAATAAAGCCTGAGTAG
GAAGGCGGCCGCTCGAGCATGCATCTAGA
6638mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACCAUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGG
G-CSF miR-CCCUGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAA
142-3p-GAAGCGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUU
seedlessCCUUUUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAU
GGAGCCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUG
CCAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUC
CCUGGGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUG
GCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCA
GGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCC
CGACGCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACA
ACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCU
GCAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUC
AGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCA
UUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCC
GUGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGC
CCUUCUUCUCUCCCUUGCACCUGUACCUCUUCCAUAAAGUAGGA
AAUGGUCUUUGAAUAAAGCCUGAGUAGGAAG
6639DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF miR-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
146aCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
CTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTAACCCATGGAATTCAGTTCTCATGGTCTTTGAATAAAGCCTG
AGTAGGAAGGCGGCCGCTCGAGCATGCATCTAGA
6640mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACCAUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGG
G-CSF miR-CCCUGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAA
146aGAAGCGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUU
CCUUUUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAU
GGAGCCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUG
CCAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUC
CCUGGGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUG
GCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCA
GGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCC
CGACGCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACA
ACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCU
GCAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUC
AGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCA
UUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCC
GUGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGC
CCUUCUUCUCUCCCUUGCACCUGUACCUCUAACCCAUGGAAUUC
AGUUCUCAUGGUCUUUGAAUAAAGCCUGAGUAGGAAG
6641DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
146a-seedCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
CTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTAGTTCTCTGGTCTTTGAATAAAGCCTGAGTAGGAAGGCGGC
CGCTCGAGCATGCATCTAGA
6642mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACCAUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGG
G-CSF-CCCUGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAA
146a-seedGAAGCGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUU
CCUUUUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAU
GGAGCCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUG
CCAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUC
CCUGGGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUG
GCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCA
GGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCC
CGACGCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACA
ACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCU
GCAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUC
AGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCA
UUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCC
GUGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGC
CCUUCUUCUCUCCCUUGCACCUGUACCUCUAGUUCUCUGGUCUU
UGAAUAAAGCCUGAGUAGGAAG
6643DNATAATACGACTCACTATA
sequenceGGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCC
having theACCATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTATGGCC
T7CTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAG
polymeraseCGACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTG
site andAAGTGTCTGGAGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCA
restrictionCTCCAAGAGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGAG
sites:GAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCCTGGGCTCCTC
G-CSF-TCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGGCAGGGTGCCTTTC
146a-CCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGCAAGCC
seedlessCTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTG
CAGCTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATG
GAGGAACTGGGGATGGCACCCGCGCTGCAGCCCACGCAGGGGGC
AATGCCGGCCTTTGCGTCCGCGTTTCAGCGCAGGGCGGGTGGAGT
CCTCGTAGCGAGCCACCTTCAATCATTTTTGGAAGTCTCGTACCGG
GTGCTGAGACATCTTGCGCAGCCGTGATAATAGGCTGCCTTCTGCG
GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTAC
CTCTAACCCATGGAATTCATGGTCTTTGAATAAAGCCTGAGTAGGA
AGGCGGCCGCTCGAGCATGCATCTAGA
6644mRNAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAGC
sequence:CACCAUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUUAUGG
G-CSF-CCCUGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAA
146a-GAAGCGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUU
seedlessCCUUUUGAAGUGUCUGGAGCAGGUGCGAAAGAUUCAGGGCGAU
GGAGCCGCACUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUG
CCAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUGGGGAUUC
CCUGGGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUUUGCAGUUG
GCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCA
GGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCC
CGACGCUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACA
ACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUGGCACCCGCGCU
GCAGCCCACGCAGGGGGCAAUGCCGGCCUUUGCGUCCGCGUUUC
AGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGCCACCUUCAAUCA
UUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCC
GUGAUAAUAGGCUGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGC
CCUUCUUCUCUCCCUUGCACCUGUACCUCUAACCCAUGGAAUUC
AUGGUCUUUGAAUAAAGCCUGAGUAGGAAG

It is likely that the binding site “seed” sequence is sufficient to induce microRNA binding, the expression of G-CSF should be down-regulated in cells transfected with miR-142-3p, miR-142-3p-seed, miR-142-5p, miR-142-5p-seed, miR-146a or miR-146a-seed. Whereas, the miR-142-3p-seedless, miR-142-5p-seedless, miR-146a-seedless should not change the expression of G-CSF, as compared with cells transfected with G-CSF mRNA without microRNA binding sites.

The binding sites for microRNAs are used in the 3′UTR of mRNA therapeutics to selectively degrade mRNA therapeutics in the immune cells to subdue unwanted immunogenic reactions caused by mRNA therapeutics delivery.

A signal-sensor polynucleotide comprising a series of 3′UTR miR binding sites which make the signal sensor polynucleotide more unstable in antigen presenting cells (APCs), such as, but not limited to mir-142-5p, mir-142-3p, mir-146a-5p and mir-146a-3p, encodes an oncology-related polypeptide of the present invention. The addition of miR binding sites in the 3′UTR making a signal sensor polynucleotide unstable would subdue modified mRNA mediated immune stimulation.

Experiments comparing the cytokine expression (e.g. TNF-alpha) induced by the signal-sensor polypeptide with APCs specific microRNA signature vs. without such signature is performed in vitro by methods described herein and/or known in the art.

Human embryonic kidney epithelial cells (HEK293A), antigen-presenting cells or cell lines with highly expressed mir-142/146, such as monocyte-derived dendritic cells (MDDC) or PBMC, are seeded at a density of 200,000 per well in 500 ul cell culture medium (InVitro GRO medium from Celsis, Chicago, Ill.). Cultured cells are transfected with G-CSF mRNAs with or without microRNA signature, as described in Example 37. The cells are transfected for five consecutive days. The transfection complexes are removed four hours after each round of transfection.

The culture supernatant is assayed for secreted G-CSF (R&D Systems, catalog #DCS50), tumor necrosis factor-alpha (TNF-alpha) and interferon alpha (IFN-alpha by ELISA every day after transfection following manufacturer's protocols. The cells are analyzed for viability using CELL TITER GLO® (Promega, catalog #G7570) 6 hrs and 18 hrs after the first round of transfection and every alternate day following that. At the same time from the harvested cells, total RNA is isolated and treated with DNASE® using the RNAEASY micro kit (catalog #74004) following the manufacturer's protocol. 100 ng of total RNA is used for cDNA synthesis using the High Capacity cDNA Reverse Transcription kit (Applied Biosystems, cat #4368814) following the manufacturer's protocol. The cDNA is then analyzed for the expression of innate immune response genes by quantitative real time PCR using SybrGreen in a Biorad CFX 384 instrument following the manufacturer's protocol.

To test the signal sensor protein expression and in vivo immune response, female BALB/C mice (n=5) are injected intramuscularly with G-CSF mRNA with or without microRNA signatures as described in Example 37. Blood is collected at 8 hours after dosing. The protein levels of G-CSF, TNF-alpha and IFN-alpha is determined by ELISA.

The difference of cytokine production is seen as measured by mouse TNF-alpha and IFN-alpha level in serum. Injection with G-CSF modified mRNA having miR-142 and miR-146a binding site or binding site seed shows a lower level of cytokine response in vivo.

Hepatocyte specific miR-122 level in rat and human primary hepatocytes was measured. Hela Cells and primary rat and human hepatocytes were cultured and RNAs were extracted from cell lysates. The miR-122 level in rat and human primary hepatocytes was compared with that in Hela cells. The miR-122 level is about 6 fold increased in primary human hepatocytes and about 12 fold increased in primary rat hepatocytes, respectively, as compared with that in Hela cells.

Primary rat and human hepatocytes and Hela cells were seeded at a density of 200,000 per well in 500 ul cell culture medium (InVitro GRO medium from Celsis, Chicago, Ill.). G-CSF mRNA having a miR-122 binding site in the 3′UTR (G-CSF miR-122-1X) (mRNA sequence is shown in SEQ ID NO: 6600; polyA tail of approximately 140 nucleotides not shown in sequence; 5′Cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU), or fully modified with pseudouridine (pU) or G-CSF mRNA with four miR-122 binding sites with the seed deleted (G-CSF no seed) (mRNA sequence is shown in SEQ ID NO: 6601; polyA tail of approximately 140 nucleotides not shown in sequence; 5′Cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU) or fully modified with pseudouridine (pU) was tested at a concentration of 250 ng per well in 24 well plates. The 24 hours after transfection, the expression of G-CSF was measured by ELISA, and the results are shown in Table 33.

TABLE 33
G-CSF mir122 expression
Primary humanPrimary rat
Hela cellsHepatocytesHepatocytes
ProteinProteinProtein
ExpressionExpressionExpression
(ng/mL)(ng/mL)(ng/mL)
G-CSF miR-122 1X167.3467.603.40
(5mC/pU)
G-CSF miR-122 1X292.18116.1825.63
(pU)
G-CSF no seed194.78129.778.39
(5mC/pU)
G-CSF no seed335.78462.8884.93
(pU)

MicroRNA control gene expression through the translational suppression and/or degradation of target messenger RNA. Mir-122 binding site containing G-CSF mRNA was translationally regulated in hepatocytes.

Primary rat and human hepatocytes and Hela cells were seeded at a density of 200,000 per well in 500 ul cell culture medium (InVitro GRO medium from Celsis, Chicago, Ill.). G-CSF mRNA (G-CSF alpha) (mRNA sequence is shown in SEQ ID NO: 6599; polyA tail of approximately 140 nucleotides not shown in sequence; 5′Cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU), G-CSF mRNA having a miR-122 binding site in the 3′UTR (G-CSF miR-122-1X) (mRNA sequence is shown in SEQ ID NO: 6600; polyA tail of approximately 140 nucleotides not shown in sequence; 5′Cap, Cap 1) fully modified with 5-methylcytidine and pseudouridine (5 mc/pU) or G-CSF mRNA with four miR-122 binding sites with the seed deleted (G-CSF no seed) (mRNA sequence is shown in SEQ ID NO: 6601; polyA tail of approximately 140 nucleotides not shown in sequence; 5′Cap, Cap1) fully modified with 5-methylcytidine and pseudouridine (5mC/pU) was tested at a concentration of 250 ng per well in 24 well plates. 24 hours after transfection, the expression of G-CSF was measured by ELISA. The G-CSF drug (mRNA) levels and protein levels are shown in Table 34.

TABLE 34
G-CSF drug and protein levels
Human HepatocytesRat Hepatocytes
DrugDrug
(mRNA)(mRNA)
level (unitProteinlevel (unitProtein
normalizedexpressionnormalizedexpression
to HPRT)(ng/ml)to HPRT)(ng/ml)
G-CSF alpha43237.6247.2626615.88784.6
(5mC/pU)
G-CSF miR-122-1X46340.974.0720171.0740.628
(5mC/pU)
G-CSF no seed70239.7298.2823170.47894.06
(5mC/pU)

The polynucleotides, primary constructs and/or mmRNA of the present invention are formulated using one of the methods described herein such as in buffer, lipid nanoparticles and PLGA. These formulations are then administered to or contacted with microphysiological systems created from organ chips as described in International Publication Nos. WO2013086502, WO2013086486 and WO2013086505, the contents of each of which are herein incorporated by reference in its entirety.

The 5′ and/or 3′ untranslated regions (UTRs) in the signal-sensor polynucleotides, primary constructs and/or mmRNA described herein may include at least one translation enhancing element (TEE). Such TEE which may be included in the 5′UTR and/or 3′UTR include, but are not limited to, those listed in Table 35, including portion and/or fragments thereof. The TEE sequence may include at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99% or more than 99% of the TEE sequences disclosed in Table 35 and/or the TEE sequence may include a 5-30 nucleotide fragment, a 5-25 nucleotide fragment, a 5-20 nucleotide fragment, a 5-15 nucleotide fragment, a 5-10 nucleotide fragment of the TEE sequences disclosed in Table 35.

TABLE 35
TEE Sequences
TEE
IdentifierSequenceSEQ ID NO
TEE-001MSCSGCNGMWA6645
TEE-002RNSGAGMGRMR6646
TEE-003RNSGAGMGRMRRR6647
TEE-004RMSCSGCNGMWR6648
TEE-005GCGAGAGAA
TEE-006GGGAGCGAA
TEE-007GCGAGAGGA
TEE-008GCGAGCGGA
TEE-009CGGAGCGAA
TEE-010CGGAGCGGA
TEE-011ACGAGAGGA
TEE-012ACGAGCGGA
TEE-013GACGAGAGGA6649
TEE-014GACGAGAGAA6650
TEE-015AGCGAGCG
TEE-016AGGAGAGGA
TEE-017GCCGAGAGA
TEE-018CGAGAGGCA
TEE-019GAGAGGAGC
TEE-020CGCGGCGGA
TEE-021CGCCGCCGC
TEE-022GCGGCTGAA
TEE-023CCGGCTGAA
TEE-024CGCCGCTGAA6651
TEE-025CGCCGCGGAA6652
TEE-026CGCCGCCGAA6653
TEE-027CCCGCGGAA
TEE-028CCCGCCGAA
TEE-029CCCGCTGAA
TEE-030CCCGGCGGA
TEE-031CGCGGCTGA
TEE-032CGGCTGCTA
TEE-033CCCGGCGGA
TEE-034AGCCGCCGCA6654
TEE-035ACGCCGCCGA6655
TEE-036GGCATTCATCGT6656
TEE-037GCATTAGTATCT6657
TEE-038TCGGTTATTGTT6658
TEE-039TCCAATTGGGAA6659
TEE-040ATCTATTGGCCA6660
TEE-041TTACTGGGTGTT6661
TEE-042AGGGTGAAGGTC6662
TEE-043GGTGGGTGTGTC6663
TEE-044CGCTTCAATGCT6664
TEE-045TGCTTCAATGCC6665
TEE-046TGTGTCTTTGCA6666
TEE-047CACGGGGACAGC6667
TEE-048AAGCTGTACATG6668
TEE-049GATGGGGGCACA6669
TEE-050ATATGTGCCCTT6670
TEE-051TCCTTCTGGGTC6671
TEE-052GGTGGGTGTGTC6672
TEE-053GAATGGATGGGG6673
TEE-054CAXGTGATATTC6674
TEE-055AGGAGGGTTTGT6675
TEE-056TGGGCGAGTGGG6676
TEE-057CGGCTCACCAGT6677
TEE-058GGTTTCXATAAC6678
TEE-059GGTGGGTGTGTC6679
TEE-060TTACTGGGTGTT6680
TEE-061AAGTCTTTGGGT6681
TEE-062CCGGCGGGU
TEE-063CCGGCGGG
TEE-064CCGGCGG
TEE-065CCGGCG
TEE-066CCGGC
TEE-067CGGCGGGU
TEE-068GGGAGACGGCGGCGGTGGCGGCGCGGGCAGAGCAAG6682
GACGCGGCGGATCCCACTCGCACAGCAGCGCACTCGG
TGCCCCGCGCAGGGTCG
TEE-069AAAGAAATGGAATCGAAGAGAATGGAAACAAATGGA6683
ATGGAATTGAATGGAATGGAATTGA
ATGGAATGGGAACG
TEE-070AAAGAAATGGAATCGAAGAGAATGGAAACAAATGGA6684
ATGGAATTGAATGGAATGGAATTGA
ATGGAATGGGAACG
TEE-071AGACAGTCAGACAATCACAAAGAAACAAGAATGAAA6685
ATGAATGAACAAAACCTTCAAGAAATATGGGATTATG
AAGAGGCCAAATGT
TEE-072AAAAGGAAATACAAGACAACAAACACAGAAACACAA6686
CCATCGGGCATCATGAAACCTCGTGAAGATAATCATCA
GGGT
TEE-073AGACCCTAATATCACAGTTAAACGAACTAGAGAAGGA6687
AGAGCAAACAAATTCAAAAGCTAGCGGAAAGCAAGA
AATAACTAAGACCAG
TEE-074AAAGACTTAAACATAAGACCTAAAACCATAAAAACCA6688
CAGAAGAAAACATAGGCAATGCCATTCAGGACATAGG
CATGGGCAAAGACTTC
TEE-075AGCAATAACCAAACAACCTCATTAAAAAGTAGGCAAA6689
GGACATAAACAGACACTTTTCAAAAGAAGACATACAC
GTGGCCAACAAACATATG
TEE-076AGAAAGAATCAAGAGGAAATGCAAGAAATCCAAAAC6690
ACTGTAACAGATATGATGAATAATGAGGTATGCACTC
ATCAGCAGACTCGACAT
TEE-077GCACTAGTCAGATCAAGACAGAAAGTCAACGAACAAA6691
GAACAGACTTAAACTACACTCTAGA
ACAAATGGACCTA
TEE-078AGCAGCCAACAAGCATATGAAATAATGCTCCACAACA6692
CTCATCATCAGAGAAATGCAAATCA
AAACCAAAAT
TEE-079AATATACGCAAATCAATAAATGTAATCCAGCATATAA6693
ACAGTACTAAAGACAAAAACCACAT
GATTATCTCAATAGATGCAGAAAAGGCC
TEE-080ATGTACACAAATCAATAAATGCAGTCCAGCATATAAA6694
CAGAACCAAACACAAAAACCACATG
ATTATCTCAATAGATGCAGAAAAGGCCTTT
TEE-081TATACCACACAAATGCAAAAGATTATTAGCAACAATT6695
ATCAACAGCAATATGTCAACAAGTT
GACAAACCTAGAGGACATGGAT
TEE-082AAACACACAAAGCAACAAAAGAACGAAGCAACAAAA6696
GCATAGATTTATTGAAATGAAAGTA
CATTCTACAGAGTGGGGGCAGGCT
TEE-083GAAATCATCATCAAACGGAATCGAATGGAATCATTGA6697
ATGGAATGGAATGGAATCATCATGG
AATGGAAACG
TEE-084AACAGAATGGAATCAAATCGAATGAAATGGAATGGAA6698
TAGAAAGGAATGGAATGAAATGGA
ATGGAAAGGATTCGAATGGAATGCAATCG
TEE-085TACAAAGAACTCAAACAAATCAGCAAGAACAAAAACA6699
ATCCCAACAAAATGTTGGACAAAG
ACATGAATAGACAATTCTCGAAAGAAGATGTACAAAT
GGCT
TEE-086TGTTGAGAGAAATTAAACAAAGCACAGATAAATGGAA6700
AAACGTGTTCATAGATTGAAAGACT
TCATGTTGTATGGTGTC
TEE-087AAACGATTGGACAGGAATGGAATCACCATCGAATGGA6701
AACGAATGGAATCTTCGAATGGAAT
TGAATGAAATTATTGAACGGAATCAAATAGAATCATC
ATTGAACAGAATCAAATTGGATCAT
TEE-088AACAATAAACAAACTCCAACTAGACACAATAGTCAAA6702
TTGCTGAAAATGAAATATAAAGGAA
CAATCTCGATGGTAGCCCAAGGA
TEE-089AAATCAATAAATGTAATTCAGCATATAAACAGAACCA6703
AAGACAAAAACCACATGATTATCTC
AATAGATGCAGAAAAGGCCTTT
TEE-090GCTCAAGGAAATAAAATAGGACACAAAGAAATGGAA6704
AAACATTCCATACTCATGGATAGAA
AGAATCAATATCATGAAATGGCC
TEE-091AACATACGCAAATCAATAAATGTAATCCAGCATATAA6705
ACAGAACCAAAGACAAAAACCACAT
GATTATCTCAATAGATGCAGAAAAGGCC
TEE-092AACAATCACTAGTCCTTAAGTAAGAGACAACACCTTTT6706
GTCACACACAGTTTGTCCTAACTTT
ATCTTGGTAATTGGGGAGACC
TEE-093AGAAAACACACAGACAACAAAAAACACAGAACGACA6707
ATGACAAAATGGCCAAGC
TEE-094ACACAACAACCAAGAAACAACCCCATTAAGAAGTGGG6708
AAAAATACATGAATAAACACATCTC
AAAAGAAGACAAACAAGTGGCTAAC
TEE-095ACAGCAGAAAACGAACATCAGAAAATCACTCTACATG6709
ATGCTTAAATACAGAGGGCAAGCAA
CCCAAGAGAAAACACCACTTCCTAAT
TEE-096GAATAGAACAGAATGGAATCAAATCGAATGAAATGGA6710
ATGGAATAGAAAGGAATGGAATGA
AATGGAATGGAAAGGATTCGAATGGAATG
TEE-097TAAGCAGAGAAAATATCAACACGAAAATAATGCAAGG6711
AGAAAAATACAGAACAATCCAAAA
TGTGGCC
TEE-098GAACAATCAATGGAAGCAGAAACAAATAAACCAAGGT6712
GTGCATCAAGGAATACATTCACGC
ATGATGGCTGTATGAGTAAAATG
TEE-099GATCAATAAATGTAATTCATCATATAAACAGAGAACT6713
AAAGACAAAAACACATGATTATCGC
AATACATGCAGAAAAGGCC
TEE-100GACAAGAGTTCAGAAAGGAAGACTACACAGAAATACG6714
CATTTTAAAGTCACTGACATGGAGA
TGACACTTAAAACCATGAACATGGATGGG
TEE-101AAGCAAAGAAAGAATGAAGCAGCAAAAGAACGAAAG6715
CAGGAATTTATTGAAAACCAAAGTA
CACTCCACAGTATGGGAGCGGACCCGAGCA
TEE-102ACCAACATAAGACAAAGAAACATCCAGCAGCTGCCTA6716
TGGCAAAAGATTACAATGTGTCAAA
CAAGAGGGCAATG
TEE-103GGACAAATTGCTAGAAATAAACAAATTACCAAAAATG6717
ATTCAAGTAGAGACAGAGAATCAA
AATAGAACTACACATAAGTGGGCCAAG
TEE-104AACATAATCCATCAAATAAACAGAACCAAAGACAAAA6718
ACCACATGATTATCTCAATAGATGC
AGAAAAGGCCTTC
TEE-105AAAATCAATATGAAAACAAACACAAGCAGACAAAGA6719
AAATTGGGCAAAAGGTTTGAGCAGA
CACTTCACCAAAGAAGTACAAATGGCAAATCAGCA
TEE-106AACCAAATTAGACAAATTGGAAATCATTACACATAAC6720
AAAAGTAATAAACTGTCAGCCTCAG
TAGTATTCATTGTACATAAACTGGCC
TEE-107AAGGAATTTAAGCAAATCAACAAGCAAAACCAAAATA6721
ATCCCATTAAAAAGTGGGTAAAGG
ACATGAATACACACTTGTCAATAGAGGACATTCAAGT
GGCCAAC
TEE-108TAACCTGATTTGCCATAATCCACGATACGCTTACAACA6722
GTGATATACAAGTTACATGAGAAAC
ACAAACATTTTGCAAGGAAACTGTGGCCAGATG
TEE-109AACTAACACAAGAACAGAAAACCAAACATCACATGTT6723
CTCACTCATAAGCGGGAGCTGAACA
ATGAGAACACACGGACACAGGGAGAGGAACATG
TEE-110TAAACTGACACAAACACAGACACACAGATACACACAT6724
ACATACAGAAATACACATTCACACA
CAGACCTGGTCTTTGGAGCCAGAGATG
TEE-111ATCAACAGACAACAGAAACAAATCCACAAAGCACTTA6725
GTTATTAGAACTGTCATACAGACTG
TACAACAACCACATTTACCAT
TEE-112AAATAAGCCAACGGTCATAAATTGCAAAGCCTTTTACA6726
ATCCAAACATGATGGAAACGATAT
GCCATTTTGAAGGTGATTTGAAAAGCACATGGTTT
TEE-113AAACAGTTCAAAAATTATTGCAACAAAATGAGAGAGA6727
TGAGTTTATCTTGCAAACTAATGGA
TGGTAGCAGTGACAGTGGCAAAACGTGGTTTGATTCT
TEE-114TAAGCAACTTCAGCAAAGTCTCAGGATACAAAATCAA6728
TGTACAAAAATCACAAGCATTCTTA
TACACCAACAACAGACAAACAAGAGTGCCAAATCATG
TEE-115AGCAAACAAACAAACAAACAAACAAACTATGACAGG6729
AACAAAACGTCACATATCAACATTA
ACAAAGAATGTAAACAGCCTAAATGCTTCACTTAAAA
GTTATAGACAGGGGCTGGGCATGGT
GGCTCACGCC
TEE-116GGAAATAACAGAGAACACAAACAAATGGGAAAACATT6730
CCATGTTCATGGATAGGAAGAATC
AATATTGTGAAAATGGCCATACT
TEE-117AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6731
TACAAAAATCACAAGCATTCTTATA
CACCAATAACAGACAAACAGAGAGCC
TEE-118AGATAAGAATAAGGCAAACATAGTAATAGGGAGTTCA6732
TGAATAACACACGGAAAGAGAACT
TACAGGGCTGTGATCAGGAAACG
TEE-119AGGAAATAAAAGAAGACACAAACAAATGGAAGAACA6733
TTCCATGCTTATGGATAGGGAGAAT
CAGTATCGTGAAAATGGCCATACT
TEE-120AACATACGAAAATCAATAAACGTAATCCAGCATATAA6734
ACAGAACCAAAGACAAAAACCACA
TGATTATCTCAATAGATGCAGAAAAGGCCTTT
TEE-121AATGGACTCGAATGAAATCATCATCAAACGGAATCGA6735
ATGGAATCATTGAATGGAATGGAAT
GGAATCATCATGGAATGGAAACG
TEE-122AAGATTTAAACATAAGACCTAAAACGACAAAAATCCT6736
AGGAGAAAACCTAAGCAATACCATT
CAGGACATAGGCATGGGCAAAGACTTCATG
TEE-123TAATGAGAAGACACAGACAACACAAAGAATCACAGAA6737
ACATGACACAGGTGACAAGAACAG
GCAAGGACCTGCAGTGCACAGGAGCC
TEE-124TAAACGTTAGACCTAAAACCATAAAAACCCTAGAAGA6738
AAACCTAGGCATTACCATTCAGGAC
ATAGGCATGGGCAAGGAC
TEE-125GAATTGAATTGAATGGAATGGAATGCAATGGAATCTA6739
ATGAAACGGAAAGGAAAGGAATGG
AATGGAATGGAATG
TEE-126GTAATGGAATGGAATGGAAAGGAATCGAAACGAAAG6740
GAATGGAGACAGATGGAATGGAATG
GAACAGAG
TEE-127AGAGAAATGCAAATCAAAACCACAATGGAATACCATC6741
TCACGCCAGTCAGAATGGCAATTAT
TAAAAAATCACAACAATTAATGATGGCAAGGCTGTGG
TEE-128AACATACACAAATCAATAAACGTAATCCAGCTTATAA6742
ACAGAACCAAAGACAAAAACCACAT
GATTATCTCAATAGATGCGGAAAAGGCC
TEE-129TAAACAGAACCAAAGACAAAAATCACATGATTATCTC6743
AATAGATGCAGAAAAGGCC
TEE-130AATGGAATGCAATCGAATGGAATGGAATCGAACGGAA6744
TGGAATAAAATGGAAGAAAACTGG
CAAGAAATGGAATCG
TEE-131AGATAAAAAGAACAGCAGCCAAAATGACAAAAGCAA6745
AAAGCAAAATCGTGTTAGAGCCAGG
TGTGGTGATGTGTGCT
TEE-132AGGAAAGTTTTCAATATGAGAAAGATACAAACCAACA6746
GAATAAGCAAACTGGATAAACAGA
AAATACAGAGAGAGCCAAGG
TEE-133GCAATCTCAGGATACAAAATCAATGTGCAAAAATCAC6747
AAGCATTCTCATACACCAATAACAG
ACAAACAGAGCCAAATCATG
TEE-134AGCATTCATATCTTGCAGTGTTGGGAAAGAGTGAGAG6748
GTTGTGATGTCAAGAAGGATAGGTC
AGAAGTGGAAGGTATGGGGGATTGTGCCTGCTGTCAT
GGCT
TEE-135AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6749
TGCAAAAATCACAAGCATTCTTATA
CACCAATAACAGACAAACAGAGAGCC
TEE-136AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6750
TGCAAAAATCACAAGCATTCTTATA
CACCAACAACAGACAAACAGAGAGCC
TEE-137TAAGCCGATAAGCAACTTCAGCAAAGTCTCAGGAGAC6751
AAAATCAATGTGCAAAAAATCACAA
GCATTCTTATACACTAATAACAGACAAACAGAGAGCC
AAATCATG
TEE-138AACGTGACATACATACAAAAAGTTTTTAGAGCAAGTG6752
AAATTTTAGCTGCTATATGTTAATTG
GTGGTAATCCC
TEE-139TACGCAAATCGATAAATGTAATCCAGCATATAAACAG6753
AACCAAAGACAAAAACCACATGATT
ATCTCAATAGATGCAGAAAAGGCC
TEE-140GCAATCGAATGGAATGGAATCGAACGGAATGGAATAA6754
AATGGAAGAAAACTGGCAAGAAAT
GGAATCG
TEE-141TTGAATCGAATGGAATCGAATGGATTGGAAAGGAATA6755
GAATGGAATGGAATGGAATTGACTC
AAATGGAATG
TEE-142TAAAGAAAAACAAACAAACAGAAATCAATGAAAATCC6756
CATTCAAAGGTCAGCAACCTCAAA
GACTGAAGGTAGATAAGCCCACAAGGATG
TEE-143GTCATATTTGGGATTTATCATCTGTTTCTATTGTTGTTG6757
TTTTAGTACACACAAAGCCACAATA
AATATTCTAGGCT
TEE-144AAAAGTACAGAAGACAACAAAAAATGAGAGAGAGAA6758
AGATAACAGACTATAGCAGCATTGG
TGATCAGAGCCACCAG
TEE-145AACCCACAAAGACAACAGAAGAAAAGACAACAGTAG6759
ACAAGGATGTCAACCACATTTTGGA
AGAGACAAGTAATCAAACACATGGCA
TEE-146AAAGACCGAAACAACAACAGAAACAGAAACAAACAA6760
CAATAAGAAAAAATGTTAAGCAAAA
CAAATGATTGCACAACTTACATGATTACTGAGTGTTCT
AATGGT
TEE-147AATCAGTAAACGTAATACAGCATATAAACAGAACCAA6761
AGACAAAAACCACATGATTATCTCA
ATAGATGCAGAAAAGGCC
TEE-148AAGCAACTTCAGCAAAGTCTCAGGACACAAAATCAAT6762
ATGCGAAAATCACAAGCATTCCTAT
ACACCAATAATAGACAAACAGAGAGCCAAATCATG
TEE-149AGCAACTTCAGCAAAATCTCAGGATACAAAATCAATG6763
TACAAAAATCACAAGCATTCTTATA
CACCAACAACAGACAAACAGAGAGCC
TEE-150TAATGCAAACTAAAACGACAATGAGATATCAATACAT6764
AACTACCAGAAAGGCTAACAAAAAA
ACAGTCATAACACACCAAAGGCTGATGAGTGAGGATG
TGCAG
TEE-151AGCAACTTCAGCAAAGTCTCAGGATACAAAATCGATG6765
TGCAAAAATCACAAGCATTCTTATA
CACCAATAACAGGCAAACAGAGAGCC
TEE-152GATATATAAACAAGAAAACAACTAATCACAACTCAAT6766
ATCAAAGTGCAATGATGGTGCAAAA
TGCAAGTATGGTGGGGACAGAGAAAGGATGC
TEE-153AAGACAGAACACTGAAACTCAACAGAGAAGTAACAAG6767
AACACCTAAGACAAGGAAGGAGAG
GGAAGGCAGGCAG
TEE-154TAAGACACATAGAAAACATAAAGCAAAATGGCAGATG6768
TAAATGCAACCTATCAATCAAAACA
TTACGAATGGCTT
TEE-155TGAAACAAATGATAATGAAAATACAACATACCAAACA6769
TACGAGATACAGTAAAAGCAGTACT
AAGATGCAAGTATATATTGCTACAAGTGCCTAC
TEE-156AATGTAATCCAGCATATAAACAGAGCCAAAGACAAAA6770
ACCACATGATTATCTCAATAGATGC
AGAAAAAGCCTTTGACAAAATTCAACAACCCTTCATGC
TAAAAACTCTCAATAAATTAGGTAT
TGATGGGACG
TEE-157ACAAAATTGATAGACCACTAGCAAGACTAATAAAGAA6771
GAAAAGAGAGAAGAATCATTACCA
TTCAGGACATAGGCATGGGCAAGGAC
TEE-158AAGGATTCGAATGGAATGCAATCGAATGGAATGGAAT6772
CGAACGGAATGGAATAAAATGGAA
GAAAACTGGCAAGAAATGGAATCG
TEE-159GATCATCAGAGAAACAGAGAAATGCAAATTAAAACCA6773
CAATGAGATACTATCTCCACACAAG
TCAGAATGGCTAT
TEE-160ATCAAAAGAAAAGCAACCTAACAAATACGGGAAGAAT6774
ATTTGAATAGACATTTCACAGGAAA
AGATATATGAATGGCCAAAAAGCAAATGAAAAG
TEE-161AACAGCAATGACAATGATCAGTAACAACAAGACTTTT6775
AACTTTGAAAAAATCAGGACC
TEE-162AAGAGCCTGAATAGCTAAAGTGATCATAAGCAAAAAG6776
AACAAAGTCGGAAGCATCACATTAC
CTGACTTCAAACTATACTCAAAGGCTATG
TEE-163ACTCAGGAAAAATAACGAATCCAACTCACAGGAGAAA6777
GAAGTACAAACCAGAAACCAATTT
CAAATTACAAGGACCAGAATACTCATGTTGGCTGGCC
AGT
TEE-164TTGACCAGAACACATTACACAATGCTAATCAACTGCAA6778
AGGAGAATATGAACAGAGAGGAGG
ACATGGATATTTTGTG
TEE-165AACATATGGAAAAAAACTCAACATCACTGATCATTAG6779
AGAAATGCAAATCAAAACCACAATG
AGATACCATCTCACGCCAGTCAGAATGGCG
TEE-166AGCAACTTCAGCAAAGACTCAGGATACAAAATCAATG6780
TGCAAAAATCACAAGCATTCTTATA
CACCAATAACAGACAGAGAGCCAAAT
TEE-167TGGGATATGGGTGAAAGAACAAGTTTGCAGAAAAGAT6781
ACAGTGAATTATGGACCATGAGTTC
GGGAAAGAAGGGTAGGACTGCG
TEE-168AGCAGTGCAAGAACAACATAACATACAAGTAAACAAA6782
CACATGGGGCCAGGTAATAAAAAG
TCAGGCTCAAGAGGTCAG
TEE-169AAGGAAAAGTAAAAGGAACTTAACACCTTCAAGAAAA6783
GACAGACAAATAACAAAACAGCAG
TTTGATAGAATGAGATATCAGGGGATGGCA
TEE-170GCTAGTTCAACATATGCAAATCAATAAACGTAATCCAT6784
CACATAAACAGAACCAATGACAAA
AACCACGATTATCTCAATAGATGCAGAAAAGGCC
TEE-171AACATCACTGATCATTAGAAACACACAAATCAAAACC6785
ACAATAAGATACCATCTAACACCAG
TCACAATGGCTATT
TEE-172AGAGCATCCACAAGGCCCAATTCAAAGAATCTGAAAT6786
AATGTATTGTTACTGCAACAGTTGTG
AGTACCAGTGGCATCAG
TEE-173GGAATAACAACAACAACAACCAAAAGACATATAGAAA6787
ACAAACAGCACGATGGCAGATGTA
AAGCCTACC
TEE-174AAACGCAGAAACAAATCAACGAAAGAACGAAGCAAT6788
GAAAGACAAAGCAACAAAAGAATG
GAGTAAGAAAGCACACTCCACAAAGTGGAAGCAGGCT
GGGACA
TEE-175AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6789
TGCAAAAATCACAAGCATTCCTATA
CACCAACAACAGACAAACAGAGAGCC
TEE-176AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6790
GGAAAAAATCACAAGCATTCCTATA
CATCAATAACAGACAAACAGAGAGCC
TEE-177ACACATTTCAAGGAAGGAAACAAGAACAGACAGAAAC6791
ACAACATACTTCATGAAACCACATT
TTAGCATCCTGGCCGAGTATTCATCA
TEE-178AGCAACTTCAGCAAAGTCTCAGGACACAAAATCAATG6792
TGCAAAAATCACAAGCATTCTTATA
CACCAATAACAGACAAACAGAGAGCC
TEE-179TATTTTACCAGATTATTCAAGCAATATATAGACAGCTT6793
AAAGCATACAAGAAGACATGTATAG
ATTTACATGCAAACACTGCACCACTTTACATAAGGGAC
TTGAGCAC
TEE-180CCCAACTTCAAATTATACTACAAGGCTACAGTAATCAA6794
AAAAGCATAGTACTATTACAAAAA
CAGACACACAGGCCAATGGAATACAAT
TEE-181AGAAAGGATTCGAATGGAATGAAAAAGAATTGAATGG6795
AATAGAACAGAATGGAATCAAATC
GAATGAAATGGAATGGAATAGAAAGGAATGGAATG
TEE-182GTTTACAGTCAAGTGTACAAACAGAATATAAGCAAAC6796
AAAAGAGAACATATACTTACAAACT
ATGCTAAGTGCCATGAAGGAAAAG
TEE-183AAGAGTATTGAAGTTGACATATCTAGACTGATCAAGA6797
ACAAAGACAAAAGGTACAGATTATC
AAGAAAATGAGCGGGCAAAGCAAGATGGCC
TEE-184AGTAGAATTGCAATTGCAAATTTCACACATATACTCAC6798
ACACAAGTACACACATCCACTTTTA
CAACTAAAAAAACTAGCACCCAGGACAGGTGCAGTGG
CT
TEE-185TGAATGCTATAGAGCAGTAAAAACAAATAAATGAACT6799
ACATTACAGCTACTTACAACCATAT
GAAAGAATATAACCATAACAATGATGAGTGGACAAAA
GCTAAGTGTGAAAGAATGCATAGT
GCTACAGCAGCCAACATTTACAGC
TEE-186GAATGGAATCAAATAGAATGGAATCGAAACAAATGGA6800
ATGGAATGGAATGGGAGCTGAGAT
TGTGTCACTGCAC
TEE-187TAAAAGTGTGCTCAACATCATTGATCATCAGAGAAATG6801
CAAATCAAAACTACAATGAGATAT
CATCTCATCCCAGTCAAAGTGGCT
TEE-188TCAGACCATAGCAGATAACATGCACATTAGCAATACG6802
ATTGCCATGACAGAGTGGTTGGTG
TEE-189ACAAACAATCCAATTCGAAAATGGGCAAGATATTTCA6803
CCAAAGACATGAGCTGATATTTCAC
TEE-190AGGAAAAACAACAACAACAACAGGAAAACAACCTCA6804
GTATGAAGACAAGTACATTGATTTAT
TCAACATTTACTGATCACTTTTCAGGTGGTAGGCAG
TEE-191AACAAAACAAAAACCCAACTCAATAACAAGAAGACAA6805
ACAACCCAATTTAAAATGAGCAAA
GAACTTGATAAACATGTCTCCAAAGAAGATACGGCCA
AAGAGCAC
TEE-192ATACAACTAAAGCAAATATAAGCAACTAAAGCAACAG6806
TACAACTAAAGCAAAACAGAACAA
GACTGCCAGGGCCTAGAAAAGCCAAGAAC
TEE-193AACAACAACAACAACAGGAAAACAACCTCAGTATGAA6807
GACAAGTACATTGATTTATTCAACA
TTTACTGATCACTTTTCAGGTGGTAGGCAGACC
TEE-194AGAGAGTATTCATCATGAGGAGTATTACTGGACAAAT6808
AATTCACAAACGAACAAACCAAAGC
GATCATCTTTGTACTGGCTGGCTA
TEE-195AGTAAATCACCATAAAGAAGGTAAGAGTTCATTCACA6809
AAAACAACAAACTGAAGAATCAGGC
CATAGTA
TEE-196AAAATAGAATGAAAGAGAATCAAATGGAATTGAATCG6810
AATGGAATCGAATGGATTGGAAAG
GAATAGAATGGAATGGAATGGAATG
TEE-197AAAAGATGCAAAAGTAGCAAATGCAATGTTAAAACAA6811
GCAAAGAAAGAATCAGGTGGACCA
CATAGTGCAGTGCTTCTC
TEE-198TTCACAGCAGCATTACGCACAATAGCCAGAAGGTGGG6812
AACAGACAAAATGCCTTTTGATGGG
TEE-199CCATAACACAATTAAAAACAACCTAAATGTCTAATAG6813
AAGAACACTGTTCAGACCGGGCATG
GTGGCTTATACC
TEE-200TGGATTTCAGATATTTAACACAAAATAGTCAAAGCAG6814
ATAAATACTAGCAACTTATTTTTAAT
GGGTAACATCATATGTTCGTGCCTT
TEE-201ATCATTGAATGCAATCACATGGAATCATCACAGAATG6815
GAATCGTACGGAATCATCATCGAAT
GGAATTGAATGGAATCATCAATTGGACTCGAATGGAA
ACATCAAATGGAATCGATTGGAAGT
GTCGAATGGACTCG
TEE-202AGAAACAGCCAGAAAACAATTATTACCTACAGCATTA6816
AAACTATTCAAATGACAGCATATTTT
TCAGCAGAAATCATGAAGGCCAGAAGGACGTGTCAT
TEE-203AAAATGATCATGAGAAAATTCAGCAACAAAACCATGA6817
AATTGCAAAGATATTACTTTTGGGA
TGGAACAGAGCTGGAAGGCAAAGAG
TEE-204AACCACTGCTCAAGGAAATAAGAGAGAACACAAACAA6818
ATGAAAAAACATTCCATGCTCATGG
ATAGGAAGAATCAG
TEE-205TACTCTCAGAAGGGAAGCAGATATTCAGCATAAATCA6819
TATTGTTTGTACAAAGAGTCTGGGCA
TGGTGAATGACACT
TEE-206TATAGTTGAATGAACACACATACACACACACATGCCA6820
CAAAACAAAAACAAAGTTATCCTCA
CACACAGGATAGAAACCAAACCAAATCCCAACACATG
GCAAGATGAT
TEE-207GCTCAAAGAAATCAGAAATGACACAAGCAAATGGAAA6821
AACATGCCATGTTCATGAATATGAA
GAATCAATATTGTTAAAATGGCCATACTGCTCA
TEE-208GGATACAAAATCAATGTACAAAAATCACAAGCATTCT6822
TATACACCAATAACAGACAAACAGA
GAGCC
TEE-209AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6823
TACAAAAATCACAAGCATTCTTATA
CACCAACAACAGACAAACAGAGAGCC
TEE-210AGGAGAATAGCAGTAGAATGACAAAATTAGATTTTCA6824
CATGAAACTTGATGACAGTGTAGGA
AATGGACTGAAAGGACAAGAC
TEE-211AGCAACTTCAGCAAAGTCTCGGGATACAAAATCAATG6825
TGCAAAAATCACAAGCATTCCTATA
CACCAATAACAGGCAAACAGAGAGCC
TEE-212AAGTTCAAACATCAGTATTAACCTTGAACATCAATGGC6826
CTACATGCATCACTTAAAACATACA
GACAGGCAAATTGGGTTAAGAAAACAAACAAGCAAAC
AAAACATGTTCCAAACATTTGTTGG
CTAT
TEE-213AAGAAACAATCAAAAGGAAGTGCTAGAAATAAAACAC6827
ACTGTAATAGAAAAGAAGAATGCC
TTATGGGCTTATCAATAGACTAGACATGGCCAGG
TEE-214AAAGAAAGACAGAGAACAAACGTAATTCAAGATGACT6828
GATTACATATCCAAGAACATTAGAT
GGTCAAAGACTTTAAGAAGGAATACATTCAAAGGCAA
AAAGTCACTTACTGATTTTGGTGGA
GTTTGCCACATGGAC
TEE-215AGCAACTTCAGCAAAGTTTCAGGATACAAAATCAATG6829
TGCAAAAATCACAAGCATTCTTATA
CACCAACAACAGACAAACAGAGAGCC
TEE-216AGAATCAAATGGAATTGAATCGAATGGAATCGAATGG6830
ATTGGAAAGGAATAGAATGGAATG
GAATGGAATG
TEE-217AAACAGAACCACAGATATCTGTAAAGGATTACACTAT6831
AGTATTCAACAGAGTATGGAACAGA
GTATAGTATTCAACAGAGTATGCAAAGAAACTAAGGC
CAGAAAG
TEE-218AAAAAATGTTCAACATCACTAGTCAGCAGAGAAATGC6832
AAATCAAAATCACAATGAGATAACT
TCTCACACCAGACAGCATGGC
TEE-219GAATCCATGTTCATAGCACAACAACCAAACAGAAGAA6833
ATCACTGTGAAATAAGAAACAAAGC
AAAACACAGATGTCGACACATGGCA
TEE-220AGGATACAAAATCAAAGTGCAAAAATCACAAGCATTC6834
TTATACACCAATAACAGACAAACAG
AGAGCC
TEE-221AACAGATTTAAACAAACCAACAAGCAAAAAACGAACA6835
ACTCCATTCAAACATGGACAAAAG
ACACGAACAGACACTTTTCAAAGAAGACATACATGTG
GCC
TEE-222AAAGACAATATACAAATGGCCAATAAGCACATGAAAA6836
GACGCTCAACATCCTTAGTCGTTAA
GGCAATGCAAATCAAAACCACAATG
TEE-223TAAACAACGAGAACACATGAACACAAAGAGGGGAAC6837
AACAGACACCAAGACCTTCTTGAGG
GTGGAGGATGGGAGGAGGGAG
TEE-224GGTTCAACTTACAATATTTTGACTTGACAACAGTGCAA6838
AAGCAATACACGATTAGTAGAAAC
ACACTTCCAATGCCCATAGGACCATTCTGC
TEE-225AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG6839
AGCAAAAATCACAAGCATTCTTACA
CACCAATAACAGACAAACAGAGAGCC
TEE-226AATCCAGCATATAAACAGAACCAAAGACAAAAACCAC6840
ATGATTATCTCAATAGATGCAGAAA
AGGCC
TEE-227TGAAAATACAAATGACCATGCAAGTAATTCCGCAGGG6841
AGAGAGCGGATATGAACAAACAGA
AGAAATCAGATGGGATAGTGCTGGCGGGAAGTCA
TEE-228GCAAATGATTATAAGTGCTGTTATAGAAACATTCAAAG6842
ACCAGAAAAGGACCACAATGGCTG
ACCAC
TEE-229AGTCAATAACAAGAAGACAAACAACCCAATTACAAAA6843
TGGGATATGAATTTAATAGATGTTA
CTCCAAGGAAGATACACAAATGGCCAAC
TEE-230ATGGTTAAAACTCAACAATGAAAACACAAACAGCGCA6844
ATTTAAAAATGGGCAAAATGACAG
GCCAGACCCAGTGGCTCATGCG
TEE-231TAACTACTCACAGAACTCAACAAAACACTATACATGC6845
ATTTACCAGTTTATTATAAAGATACA
AGTCAGGAACAGCCAAATGGAAGAAATGTAAATGGCA
AG
TEE-232AACAGACCATAAATAAACACAGAAGACACACGAGTGT6846
AAAGTCAGTGCCCCGCTGCGAATTA
AATCGGGGTGATGTGATGGCGAGTGAGTGGGTAGTT
TEE-233GAATAGAATAGAATGGAATCATCGAATGGAATCGAAT6847
GGAATCATCATGATATGGAATTGAG
TGGAATC
TEE-234GGAATCTATAATACAGCTGTTTATAGCCAAGCACTAAA6848
TCATATGATACAGAAAACAAATGC
AGATGGTTTGAAGGGTGGG
TEE-235AAGATAGAGTTGAAACAGTGGACAATTAAAGAGTAAT6849
TTGGAAGAATGGTGAAATTACAGCC
ATGCTTTGAATCAGGCGGGTTCACTGGC
TEE-236TGAAAAGAAGAATGACCATAAGCAAGCAGATGAAAA6850
ACAAAACAGAATTTTTACAGACGTCT
TGGACTGATATCTTGGGC
TEE-237AGGAATCTATAATACAGCTGTTTATAGCCAAGCACTAA6851
ATCATATGATACAGAAAACAAATG
CAGATGGTTTGAAGGGTGGG
TEE-238AGGAAAAGAAAGAAATAGAAAATGCGAAATGGTAAG6852
AAAAAACAGCATAATAAACATTTGT
ATGGTGTTGATGGACAATGCATT
TEE-239TAACAGTACCAAAAAACAGTCATAATCTTCAAGAGCTT6853
AAATTTAGCATGAAAGGAAGACAT
TCATCAAAGAATCACACAAAGGAATGTAAAATTAAAT
GGAGATTAGTGCCAGGAAAGAGC
TEE-240GCAAAACACAAACAACGCCATAAAAAACTGGGCAAAG6854
GATATGAACAGACATTTTTCAAAAC
AAAACATACTTATGGCCAAC
TEE-241AACAAAATTGAACAACATGCAAAGAAACATAAACGAA6855
GCAATGAAAGTGTGCAGATCCACT
GAAATGAAAGTGCTGTCCAGAGTGGGAGCCAGCTCGA
GA
TEE-242GAATGGAATCAACATCAAACGGAATCAAACGGAATTA6856
TCGAATGGAATCGAAGAGAATCATC
GAATGGCCACGAATGGAATCATCTAATGGAATGGAAT
GGAATAATCCATGG
TEE-243TACAAGAAAATCACAGTAACATTTATAAAACACAGAA6857
GTGTGAACACACAGCTATTGACCTT
GAAAACAGTGAAAGAGGGTCAGCTGTAGAACTAAGAC
ATAAGCAAAGTTTTTCAATCAAGAA
TACATGGGTGGCC
TEE-244AAGAATTGGACAAAACACACAAACAAAGCAAGGAAG6858
GAATGAAAGGATTTGTTGAAAATGA
AAGTACACTCCACAGTGTGGGAGCAG
TEE-245ACAGTTAACAAAAACCGAACAATCTAATTACGAAATG6859
AACAAAAGATATGAACAGACATTTC
ACCCGAGAGTATACAGGGGCCAGGCATGGT
TEE-246AAACGCACAAACAAAGCAAGGAAAGAATGAAGCAAC6860
AAAAGCAGAGATTTATTGAAAATGA
AAAATACACTCCACAGGGTGGG
TEE-247CACCATGAGTCATTAGGTAAATGCAAATCAAAACCAC6861
AATGAAATACTTCACACCCATGAAG
ATGGCTATAATAAAAAAACAGACA
TEE-248AGCAACTTCAGCAAAGTCTCAGGAGACAAAATCAATG6862
TACAAAAATCACAAGCATTCTTATA
CACCAATAACAGACAAACAGAGAGCC
TEE-249TGACATGCAAGAAATAAGGAAGTGCAAAAACAAACAA6863
ACAAACAACAACAACAACAACAAC
AACAACAACAAAAAACAGTCCCAAAAGGATGGGCAG
TEE-250AGACTTGAAAAGCACAGACAACGAAAGCAAAAATGG6864
ACAAATGGAATCACATCAAGCTAAA
AGGTTTTGCATGGCAAAGG
TEE-251GCAAAAGAAACAATCAGTAGAGTAAACAGACAACTCA6865
TAGAATGCAAGAAAATCATCGCAA
TCTGTACATCCAACAAAGGGCT
TEE-252ACAAAATCAAACTAACCTCGATAAGAATGCAAGTGAA6866
TCAAAATGAGTTTCAAGGGGTTGTG
GCTAGTACACGCTTTCTACAGCTG
TEE-253ACAAACCACTGCTCAAGGAAATAAGGACACAAACAAA6867
TGGAACAACATTCCGTGCTCATGGA
TAGGAAGAATCAATATCGTGAAAATGGCCATACT
TEE-254GAACGATTTATCACTGAAAATTAATACTCATGCAAGTA6868
GTAAACGAATGTAATGACCATGAT
AAGGAGACGGACGGTGGTGATAGT
TEE-255AGCAGAAGAAATAACTGAAATCAGAGTGAAACTGAAT6869
CAAATTGAGATGCAAAAATACATA
CGAAATGGCCAG
TEE-256TGAATAGACACACAGACCAATGGAACAGAATAGAGAA6870
CACAGAATAAATCTGCACACTTATA
GCCAGCTGATTTTTGACAAATTTGCCAAG
TEE-257AGCAACTTCAGCAGTCTCAGTATACAAAAACAATGTG6871
CAAAAATCACAAGCATTCCTATATG
CCAATAACAGACAAACAGAGAGCC
TEE-258ACCAATCAAGAAAACAATGCAACCCACAGAGAATGGA6872
CAAAAGCAAGGCAGGACAATGGCT
TEE-259GCCACAATTTTGAAACAACCATAATAATGAGAATACA6873
CAAGACAACTCCAATAATGTGGGAA
GACAAACTTTGCAATTCACATCATGGC
TEE-260GAAAATGAACAATATGAACAAACAAACAAAATTACTA6874
CCCTTACGAAAGTACGTGCATTCTA
GTATGGTGACAAAAAGGAAA
TEE-261TATGCAAATCAATAAACATAATCCATCACATAAACAG6875
AAACAAAGACAAAATGACATGATTA
TCTCAATAGATGCAGAAAAGGCC
TEE-262CACCCATCTGTAGGACCAGGAAGCCTGATGTGGGAGA6876
GAACAGCAGGCTAAATCCAGGGTTG
GTCTCTACAGCAGAGGGAATCACAAGCCTGTTAGCAA
GTGAAGAACCAACACTGGCAAGAGT
GTGAAGGCC
TEE-263AGGATACAAAATCAATGTACAAAAATCACAAACATTC6877
TTATACACCAACAACAGACAAACAG
AGAGCCAAATCATGGGTG
TEE-264AGGAAAATGCAAATCAGAACGACTATAACACACCATC6878
TCAAACTCGTTAGGATGGCTATTAT
CAAAAAGTCAAGAGATAACAAATGTGGGCAAGGG
TEE-265GTAACAAAACAGACTCATAGACCAATAGAACAGAATA6879
GAGAATTCAGAAATAAGACTGCACT
TCTATGACCATGTGATCTTAGACAAACCT
TEE-266AAAGGAAAACTACAAAACACTGCTGAAAGAAATCATT6880
GACAACACAAACAAATGGAAACAC
ATCCCAAGATCATGGGTGGGTGGAATCAAT
TEE-267ACACACATACCAACAGAACATGACAAAAGAACAAAAC6881
CAGCCGCATGCATACTCGATGGAG
ACAAAGGTAACACTGCAGAATGGTGAAGGAAGAACAG
TCATTTTAATGACAGTGTTGGCT
TEE-268AACTAAGACAACAGATTGATTTACACTACTATTTTCAC6882
ACAGCCAAAAATATCACTATGGCAA
TCGTCAAAAGGTCAATTCAAAGATGGGACAGT
TEE-269GATCAGCTTAGAATACAATGGAACAGAACAGATTAGA6883
ACAATGTGATTTTATTAGGGGCCAC
AGCACTGTTGACTCAAGTACAAGTTCTGACTCATGTAG
AACTAACACTTTT
TEE-270GAATGGAATCAAATCGAATGAAATGGAATGGAATAGA6884
AAGGAATGGAATGAAATGGAATGG
AAAGGATTCGAAT
TEE-271AAATGAACAAAACTAGAGGAATGACATTACCTGACTT6885
CAAATTATACTACAGAGCTATAGTA
ACCAAAACAGCATGGTACAGGCAT
TEE-272GGACAACATACACAAATCAGTCAAGATACATCATTTC6886
AACAGAATGAAAGACAAAAACCATT
TGATCACTTCAATCGATGATGAAAAAGCA
TEE-273AACTTCAGCAAATTCTCAGGATACAAAATCAATGTGCA6887
AAAACCACAAGCATTCCTATACAC
CAATAATAGACAGTGAGCCAAAT
TEE-274TATGACTTTCACAAATTACAGAAAAAGACACCCATTTG6888
ACAAGGGAACTGAAGGTGGTGAAG
ACATACTGGCAGGCTAC
TEE-275AACAGCAATAGACACAAAGTCAGCACTTACAGTACAA6889
AAACTAATGGCAAAAGCACATGAA
GTGGGACAT
TEE-276TGTAACACTGCAAACCATAAAAACCGTAGAAGAAAAC6890
CTAGACAATACTATTCAGGACATAG
GCATGGGCAAAGAC
TEE-277GAAGAAGAAAAAACATGGATATACAATGTCAACAGAA6891
ATCAAGGAGAAACGGAATTTCACC
AATCAATTTAGTGATCTGGGTT
TEE-278AAAACACACAAACATACATGTGGATGCACATATAAAC6892
ATGCACATACACACACACATAAATG
CACAAACACACTTAACACAAGCACACATGCAAACAAA
CACATGG
TEE-279TAGAAGGAATTTGATACATGCTCAGAAATACAGGCAA6893
AGGAAGTAGGTGCCTGCCAGTGAAC
ACAGGGGAACTATGGCTCCTA
TEE-280TGACTAAACAGAGTTGAACAAGAACAAAAAGCAAATT6894
TGCAGAAATGAAATACATACTAATT
GAAAGTCCATGGACAGGCTCAACAGATGATATAGATA
CAGCTAAAGAGATAATTAGTGAAAT
GGATCAG
TEE-281AAGTAATAAGACTGAATTAGTAATACAAAGTGTCTCA6895
ACAAAGAAAATTGCGGGACTGTTCA
TGCTCATGGACAGGAAGAATCAATATCATGAAAATGG
CC
TEE-282ACAGACAGAGATTTAAAACAATAAACAAGCAGTAAGC6896
AAACACAGATAACAAAATGACATG
ATCCAACAAATACTCAGAAGGAGACTTAGAAATGAAT
TGAGGGTC
TEE-283AGAAAAAAACAAACAGCCCATTAAAAGGTAGACAAA6897
GGACATGAACACTTTTCAAAAGAAG
ACATACATGTGGCCAAACAGCATG
TEE-284AAAAATGACCAGAGCAATAGAATGCATTGACCAGATA6898
AAGACCTTCACGTATGTTGAACTAA
AATGTGTGGTGCAGGTG
TEE-285AATCAGTCTAGATCTTAAAGGAACACCAGAGGGAGTA6899
TTTAAATGTGCCCAATAAGCAAGAA
TTATGGTGATGTGGAAGTA
TEE-286GAATGGAATGGAAAGGAATCGAAACGAAAGGAATGG6900
AGACAGATGGAATGGAATGGAACAG
AGAGCAATGG
TEE-287GGAATGGAATGAACACGAATGTAATGCAACCCAATAG6901
AATGGAATCGAATGGCATGGAATAT
AAAGAAATGGAATCGAAGAGAATGGAAACAAATGGA
ATGGAATTG
TEE-288AGGACATGAATAGACAATTCTCAAAAGAAGATACACA6902
AGTGGCAAACAAACACATGAAAAA
AGACTCAACATTAGTAATGACCATGGAAATGCAAATC
TEE-289TCCAGTCGATCATCATATAGTCAGCACTTATCATACAC6903
CAAGCCGTGTGCAAGGAAAGGGAA
TACAACCATGAACATGATAGATGGATGGTT
TEE-290TACAGATAAGAAAATTGAGACTCAAGAGTATTACATA6904
AATTGTTTCAGCTACCACAGCAAAA
AATGGTATGGTTGGGAATCAAGCTCAGGG
TEE-291AGCCTATCAAAAAGTGGGCTAAGAATATGAATACACA6905
ATTCTCAAAAGAAGATATACAAATG
GGCAACAAACATATGAAAACATACTCAACATCACTAA
TGATCAGGGAAATG
TEE-292GAAAATGAACAATATGAACAAACAAACAAAATTACTA6906
CCCTTACGAAAGTACGTGCATTCTA
GTATGGTGACAAAAAGGAAAG
TEE-293ACATACGCAAATCAATAAACATAATCCATCACATAAA6907
CAGAACCAAAGACAAAAATCACATG
ATTATCTCAATAGATGCAGAAAAGGCCTTCGAC
TEE-294AAGAGTATCAACAGTAAATTACATTAGCAGAAGAATC6908
AACAAACATGAAAATAGAAATTATG
GTAGCCAAAGAACAG
TEE-295AATCGAATGGAATCAACATCAAACGGAAAAAAACGGA6909
ATTATCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-296GAAAGGAATAGAATGGAATGGATCGTTATGGAAAGAC6910
ATCGAATGGGATGGAATTGACTCGAATGGATTGGACT
GGAATG
GAACGGACTCGAATGGAATGGACTGGAATG
TEE-297TAAGCAATTTCAGCAGTCTCAGGATACAAAATCAATGT6911
GCAAAAATCACAAGCATTCTTATACACCAACAACAGA
CAAAC
AGAGAGCCAAATCG
TEE-298AACGGAATCAAACGGAATTATCGAATGGAATCGAAGA6912
GAATCATCGAATGGCCACGAATGGAATCATCTAATGG
AATGG
AATGGAATAATCCATGGACCCGAATG
TEE-299ACATCAAACGGAATCAAACGGAATTATCGAATGGAAT6913
CGAAAAGAATCATCGAACGGACTCGAATGGAATCATC
TAATGG
AATGGAATGGAAG
TEE-300ATCGAATGGAATCAACATCAAACGGAAAAAAACGGAA6914
TTATCAAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-301GAATAATCATTGAACGGAATCGAATGGAAACATCATC6915
GAATGGAAACGAATGGAATCATCATCGAATGGAAATG
AAAGG
AGTCATC
TEE-302CATCAAACGGAATCAAACGGAATTATCGAATGGAATC6916
GAAAAGAATCATCGAACGGACTCGAATGGAATCATCT
AATGGA
ATGGAATGGAAGAATCCATGGACTCGAATG
TEE-303AAACGGAATCAAACGGAATTATCGAATGGAATCGAAG6917
AGAATCATCGAATGGACTCGAATGGAATCATCTAATG
GAATGG
AATGGAAGAATCCATGG
TEE-304ATACACAAATCAATAAATGTAATCCAGCATATAAACA6918
GAACCAAAGACAAAAACCATATGATTATCTCAATGGA
TGCAGA
AAAGGCC
TEE-305AATCGAATAGAATCATCGAATGGACTCGAATGGAATC6919
ATCGAATGTAATGATGGAACAGTC
TEE-306TGGAATGGAATCATCGCATAGAATCGAATGGAATTAC6920
CATCGAATGGGATCGAATGGTATCAACATCAAACGCA
AAAAAA
CGGAATTATCGAATGGAATCGAAGAGAATCTTCGAAC
GGACCCG
TEE-307ATGGAATGGAATGGAATGGAATTAAATGGAATGGAAA6921
GGAATGGAATCGAATGGAAAGGAATC
TEE-308GTCGAAATGAATAGAATGCAATCATCATCAAATGGAA6922
TCCAATGGAATCATCATCAAATAGAATCGAATGGAAT
CATCAA
ATGGAATCGAATGGAGTCATTG
TEE-309TGGAATTATCGAAAGCAAACGAATAGAATCATCGAAT6923
GGACTCGAATGGAATCATCGAATGGAATGGAATGGAA
CAG
TEE-310AAAGGAATGGAATGCAATGGAATGCAATGGAATGCAC6924
AGGAATGGAATGGAATGGAATGGAAAGGAATG
TEE-311AATCTAATGGAATCAACATCAAACGGAAAAAAACGGA6925
ATTATCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-312TACACAACAAAAGAAATACTCAACACAGTAAACAGAC6926
AACCTTCAGAACAGGAGAAAATATTTGCAAATACATC
TAACAA
AGGGCTAATATCCAGAATCT
TEE-313TGCAATCCTAGTCTCAGATAAAACAGACATTAAACCA6927
ACAAAGATCAAAAGAGACAAAGAAGGCCATTAC
TEE-314GAATCGAATGGAATCAACATCAAACGGAAAAAAACGG6928
AATTATCGAATGGAATCGAAAAGAATCATCGAATGGA
CC
TEE-315AATGGAATCGAATGGAATGCAATCCAATGGAATGGAA6929
TGCAATGCAATGGAATGGAATCGAACGGAATGCAGTG
GAAGG
GAATGG
TEE-316GAACACAGAAAAATTTCAAAGGAATAATCAACAGGGA6930
TTGATAACTAACTGGATTTAGAGAGCCAAGGCAAAGA
GAATC
AAAGCACAGGGCCTGAGTCGGAG
TEE-317AGTTGAATAGAACCAATCCGAATGAAATGGAATGGAA6931
TGGAACGGAATGGAATTGAATGGAATGGAATGGAATG
CAATG
GA
TEE-318AACTCGATTGCAATGGAATGTAATGTAATGGAATGGA6932
ATGGAATTAACGCGAATAGAATGGAATGGAATGTAAT
GGAACG
GAATGGAATG
TEE-319AAGCGGAATAGAATTGAATCATCATTGAATGGAATCG6933
AGTAGAATCATTGAAATCGAATGGAATCATAGAATGG
AATCCA
AT
TEE-320AATGGAATCGAAAGGAATAGAATGGAATGGATCGTTA6934
TGGAAAGATATCGAATGGAATGGAATTGACTCGAATG
GAATG
GACTGGAATGGAACG
TEE-321TAACGGAATAATCATCGAACAGAATCAAATGGAATCA6935
TCATTGAATGGAATTGAATGGAATCTTCGAATAGACAT
GAATG
GACCATCATCG
TEE-322AACGGAATCAAACGGAATTATCGAATGGAATCGAATA6936
GAATCATCGAACGGACTCGAATGGAATCATCTAATGG
AATGGA
ATGGAAG
TEE-323ATTGGAATGGAACGGAACAGAACGGAATGGAATGGAA6937
TAGAATGGAATGGAATGGAATGGTATGGAATGGAATG
GAATG
GTACG
TEE-324AATCCACAAAGACAACAGAAGAAAAGACAACAGTAG6938
ACAAGGATGTCAACCACATTTTGGAAGAGACAAGTAA
TCAAAC
ACATGGCA
TEE-325GAATCGAATGGAATCAACATCAAACGGAAAAAAACGG6939
AATTATCGAATGGAATCGAAAAGAATCATCGAACGGA
CTCGA
ATGGAATCATCTAATGGAATGGAATGGAAGAATCCAT
GG
TEE-326AATGGAATCGAATGGAATCATCATCAAATGGAATCTA6940
ATGGAATCATTGAACGGAATTGGATGGAATCGTCAT
TEE-327CAACATCAAACGGAAAAAAACGGAATTATCGAATGGA6941
ATCGAAGAGAATCATCGAATGGACC
TEE-328CACAACCAAAGCAATGAAAGAAAAGCACAGACTTATT6942
GAAATGAAAGTACACACCACAGAATGGGAGCAGGCTC
AAGCA
AGC
TEE-329ATCAAAGGGAATCAAGCGGAATTATCGAATGGAATCG6943
AAGAGAATCATCGAATGGACTCGAATGGAATCATGTG
ATGGA
ATGGAATGGAATAATCCACGGACT
TEE-330GGAATCGAATGGAATCAATATCAAACGGAGAAAAACG6944
GAATTATCGAATGGAATCGAAGAGAATCATCGAATGG
ACC
TEE-331AGGAATGGACACGAACGGAATGCAATCGAATGGAATG6945
GAATCTAATAGAAAGGAATTGAATGAAATGGACTGG
TEE-332GGAAGGGAATCAAATGCAACAGAATGTAATGGAATGG6946
AATGCAATGGAATGCAATGGAATGGAATGGAATGCAA
TGGAA
TGG
TEE-333AAATTGGATTGAATCGAATCGAATGGAAAAAATGAAA6947
TCAAATGAAATTGAATGGAATCGAAATGAATGTAAAC
AATGG
AATCCAATGGAATCCAATGGAATCGAATCAAATGGTTT
TGAGTGGCGTAAAATG
TEE-334AATGGAAGGGAATGGAATGGAATCGAATCGAATGGAA6948
CAGAATTCAATGGAATGGAATGGAATGGAATGGAATC
GAATG
GAATGG
TEE-335GAAAAATCATTGAACGGAATCGAATGGAATCATCATC6949
GGATGGAAACGAATGGAATCATCATCGAATGGAAATG
AAAGG
AGTCATC
TEE-336GGAATCGAATGGAATCAACATCAAACGGAGAAAAACG6950
GAATTATCGAATGGAATCGAAGAGAATCATCGAATGG
ACC
TEE-337AAAGAAATGTCACTGCGTATACACACACACGCACATA6951
CACACACCATGGAATACTACTCAGCTATACAAAGGAA
TGAAAT
AATCCACAGCCAC
TEE-338GGAATCGAATGGAATCAATATCAAACGGAAAAAAACG6952
GAATTATCGAATGGAATCGAAGAGAATCATCGAATGG
ACC
TEE-339TGAACGGAATCGAATGGAATCATCATCGGATGGAAAC6953
GAATGGAATCATCATCGAATGGAAATGAAAGGAGTCA
TC
TEE-340GAATAGAACGAAATGGAATGGAATGGAATGGAATGGA6954
AAGGAATGGAATGGAATGGAACG
TEE-341TGGAATTATCGTCGAATAGAATCGAATGGTATCAACAT6955
CAAACGGAAAAAAACGGAATTATCGAATGGAATCGAA
GAGA
ATCATCGAACGGACTCGAATGGAATCATCTAATGGAA
TGGAATGGAATAATCCATGG
TEE-342GACAAAAAGAATCATCATCGAATAGAATCAAATGGAA6956
TCTTTGAATGGACTCAAAAGGAATATCGTCAAATGGA
ATCAAA
AGCCATCATCGAATGGACTGAAATGGAATTATCAAAT
GGACTCG
TEE-343AACCAAACCAAGCAAACAAACAAACAGTAAAAACTCA6957
ATAACAACCAACAAACAGGAAATACCAGGTAATTCAG
ATTAT
CTAGTTATGTGCCATAGT
TEE-344GAATGAATTGAATGCAAACATCGAATGGTCTCGAATG6958
GAATCATCTTCAAATGGAATGGAATGGAATCATCGCAT
AGAAT
CGAATGGAATTATCAACGAATGGAATCGAATGGAATC
ATCATCAGATGGAAATGAATGGAATCGTCAT
TEE-345TGGAATGGAATCAAATCGCATGGAATCGAATGGAATA6959
GAAAAGAATCAAACAGAGTGGAATGGAATGGAATGG
AATGGA
ATCATGCCGAATGGAATG
TEE-346AAATGGAATAATGAAATGGAATCGAACGGAATCATCA6960
TCAAAAGGAACCGAATGAAGTCATTGAATGGAATCAA
AGGCA
ATCATGGTCGAATGGAATCAAATGGAAACAGCATTGA
ATAGAATTGAATGGAGTCATCACATGGAATCG
TEE-347GAATTAACCCGAATAGAATGGAATGGAATGGAATGGA6961
ACAGAACGGAACGGAATGGAATGGAATGGAATGGAAT
GGAATG
TEE-348AAGATATACAAGCAGCCAACAAACATACGAAAGAATG6962
CTCAACATCACTAATCCTCAGAGAAATTTAAATCAAAA
CCACA
ATGAGTTACAATCTCATACCAGTCAGAAT
TEE-349AGATAAGTGGATGAACAGATGGACAGATGGATGGATG6963
GATGGATGGATGGATGGATGCCTGGAAGAAAGAAGAA
TGGAT
AGTAAGCTGGGTATA
TEE-350AGAATTACAAACCACTGCTCAACAAAATAAAAGAGTA6964
CACAAACAAATGGAAGAATATTCCATGCTTATGGATA
GGAAGA
ATCAATATTGTGAAAATGGCCATACT
TEE-351CATCGAATGGACTCGAATGGAATAATCATTGAACGGA6965
ATCGAAGGGAATCATCATCGGATGGAAACGAATGGAA
TCATCA
TCGAATGGAAATG
TEE-352AAAGGAATCAAACGGAATTATCGAATGGAATCGAAAA6966
GAATCATCGAACGGACTCGAATGGAATCATCTAATGG
AATGG
AATGGAAGAATCCATGGACTCGAATG
TEE-353GGATATAAACAAGAAAACAACTAATCACAACTCAATA6967
TCAAAGTGCAATGATGGTGCAAAATGCAAGTATGGTG
GGGAC
AGAGAAAGGATGC
TEE-354AACATCAAACGGAAAAAAACGGAAATATCGAATGGAA6968
TCGAAGAGAATCATCGAATGGACC
TEE-355TAAAATGGAATCGAATGGAATCAACATCAAATGGAAT6969
CAAATGGAATCATTGAACGGAATTGAATGGAATCGTC
AT
TEE-356AATCATCATCGAATGGAATCGAATGGTATCATTGAATG6970
GAATCGAATGGAATCATCATCAGATGGAAATGAATGG
AATCG
TCAT
TEE-357CAATGCGTCAAGCTCAGACGTGCCTCACTACGGCAATG6971
CGTCAAGCTCAGGCGTGCCTCACTAT
TEE-358TAAGCTGATAAGCAACTTTAGCAAAGTCTCAGGATAC6972
AAAATCAATGTACAAAAATCACAAGCATTCTTATACAC
CAACA
ACAGACAGACGGAGAGCCAAA
TEE-359AATCAAAGAATTGAATCGAATGGAATCATCTAATGTA6973
CTCGAATGGAATCACCAT
TEE-360ATGAACACGAATGTAATGCAATCCAATAGAATGGAAT6974
CGAATGGCATGGAATATAAAGAAATGGAATCGAAGAG
AATGG
AAACAAATGGAATGGAATTGAATGGAATGGAATTG
TEE-361ATCAAACGGAATCAAACGGAATTATCGAATGGAATCG6975
AAGAGAATCATCGAACGGACTCGAATGGAATCATCTA
ATGGAA
TGGGATGG
TEE-362AATGGAAAGGAATCAAATGGAATATAATGGAATGCAA6976
TGGACTCGAATGGAATGGAATGGAATGGACCCAAATG
GAATG
GAATGGAATGGAATG
TEE-363GGAATACAACGGAATGGAATCGAAAAAAATGGAAAG6977
GAATGAAATGAATGGAATGGAATGGAATGGAATGGAT
GGGAA
TGGAATGGAATGG
TEE-364GAATCAAGCGGAATTATCGAATGGAATCGAAGAGAAT6978
CATCGAAAGGACTCGAATGGAATCATCTAATGGAATG
GAATG
GAATAATACACGGACC
TEE-365AAGATAACCTGTGCCCAGGAGAAAAACAATCAATGGC6979
AACAAAAGCAGAAACAACACAAATGATACAATTAGCA
GACAG
AAACATTGAGATTGCTATT
TEE-366AATGGACTCCAATGGAATAATCATTGAACGGAATCTA6980
ATGGAATCATCATCGGATGGAAATGAGTGGAATCATC
ATCGAA
TGGAATCG
TEE-367AATCTATAAACGTAATCCATCACATAAACAGGACCAA6981
AGAGAAAAACCGCATGATTATCTCAAGAATGCAGAAA
AGGCC
TEE-368TAATTGATTCGAAATTAATGGAATTGAATGGAATGCAA6982
TCAAATGGAATGGAATGTAATGCAATGGAATGTAATA
GAATG
GAAAGCAATGGAATG
TEE-369AAAGGAATGGACTTGAACAAAATGAAATCGAACGATA6983
GGAATCGTACAGAACGGAAAGAAATGGAACGGAATG
GAATG
TEE-370TGAGCAGGGAACAATGCGGATAAATTTCACAAATACA6984
ATGTTGAGCAAAAGAAAGACACAAAAGAATACACACA
TACAC
ACCATATGGGCTAGG
TEE-371AATGGAATCGAACGGAATCATCATCAAACGGAACCGA6985
ATGGAATCATTGAATGGAATCAAAGGCAATCATGGTC
GAATG
TEE-372AATGGAATGGAATGTACAAGAAAGGAATGGAATGAAA6986
CCGAATGGAATGGAATGGACGCAAAATGAATGGAATG
GAAGT
CAATGG
TEE-373AACGGAAAAAAACGGAATTATCGAATGGAATCGAAGA6987
GAATCATCGAATGGACC
TEE-374GGAATAATCATTGAACGGAATCGAATGGAATCATCAT6988
CGGATGGAAACGAATGGAATCATCATCGAATGGAAAT
GAAAG
GAGTCATC
TEE-375GGAACGAAATCGAATGGAACGGAATAGAATAGACTCG6989
AATGTAATGGATTGCTATGTAATTGATTCGAATGGAAT
GGAAT
CG
TEE-376TGAAAGGAATAGACTGGAACAAAATGAAATCGAATGG6990
TAGGAATCATACAGAACAGAAAGAAATGGAACGGAAT
GGAATG
TEE-377AACCCGAATAGAATGGAATGGAATGGAATGGAACGGA6991
ACGGAATGGAATGGAATGGATTGGAATGGAATGGAATG
TEE-378AAAGAGAATCAAATGGAATTGAATCGAATGGAATCGA6992
ATGGATTGGAAAGGAATAGAATGGAATGGAATGGAAT
GGAAT
GGAATGGAATG
TEE-379AATGGAATCATCAGTAATGGAATGGAAAGGAATGGAA6993
AGGACTGGAATGGAATGGAATGGAATGGAATGG
TEE-380GGAACAAAATGAAATCGAACGGTAGGAATCGTACAGA6994
ACGGAAAGAAATGGAACGGAATGGAATGCACTCAAAT
GGAAA
GGAGTCCAATGGAATCGAAAGGAATAGAATGGAATGG
TEE-381AGAATGAGATCAAGCAGTATAATAAAGGAAGAAGTAG6995
CAAAATTACAACAGAGCAGTGAAATGGATATGCTTTCT
GGCA
ATAATTGTGAAAGGTCTGGTAATGAGAAAGTAGCAAC
AGCTAGTGGCTGCCAC
TEE-382AACAAATGGAATCAACATCGAATGGAATCGAATGGAA6996
ACACCATCGAATTGAAACGAATGGAATTATCATGAAA
TTGAAA
TGGATGGACTCATCATCG
TEE-383TAACATGCAGCATGCACACACGAATACACAACACACA6997
AACATGTATGCACGCACACGTGAATACACAACACACA
CAAACA
TGCATGCATGCATACATGAATACACAGCACACAAATA
TCCAGCAT
TEE-384GAATGGAATCAACATCAAACGGAAAAAAAACGGAATT6998
ATCGAATGGAATCGAATAGAATCATCGAATGGACC
TEE-385AATCGAATGAAATGGAGTCAAAAGGAATGGAATCGAA6999
TGGCAAGAAATCGAATGTAATGGAATCGCAAGGAATT
GATGT
GAACGGAACGGAATGGAAT
TEE-386AATGGAATTGAACGGAAACATCAGCGAATGGAATCGA7000
AAGGAATCATCATGGAATAGATTCGAATGGAATGGAA
AGGAA
TGGAATGGAATG
TEE-387ATGGAATCAACATCAAACAGAATCAAACGGAATTATC7001
GAATGGAATCGAAGACAATCATCGAATGGACTCGAAT
GGAATC
ATCTAATGGAATGGAATGGAAGAATCCATGGTCTCGA
ATGCAATCATCATCG
TEE-388GAATAATCATTGAACGGAATCGAATGGAATCATCTTCG7002
GATGGAAACGAATGGAATCATCATCGAATGGAAATGA
AAGGA
GTCATC
TEE-389AATGGACTCGAATGGAATAATCATTGAACGGAATCGA7003
ATGGAATCATCATCGGATGGAAATGAGTGGAATCATC
ATCGAA
TGGAATCG
TEE-390AAATGAAATCGAACGGTAGGAATCGTACAGAACGGAA7004
AGAAATGGAACGGAATGGAATGCAATCGAATGGAAAG
GAGTC
CAATGGAAGGGAATCGAAT
TEE-391TACCAAACATTTAAAGAACAAATATCAATCCTACGCA7005
AACCATTCTGAAACACAGAGATGGAGGATATACAGCG
AAACTC
ATTCTACATGGCC
TEE-392TATTGGAATGGAATGGAATGGAGTCGAATGGAACGGA7006
ATGCACTCGAATGGAAGGCAATGCAATGGAATGCACT
CAACA
GGAATAGAATGGAATGGAATGGAATGG
TEE-393GGAATTTAATAGAATGTACCCGAATGGAACGGAATGG7007
AATGGAATTGTATGGCATGGAATGGAA
TEE-394GCAATCCAATAGAATGGAATCGAATGGCATGGAATAT7008
AAAGAAATGGAATCGAAGAGAATGGAGACAAATGGA
ATGGAA
TTGAATGGAATGGAATTG
TEE-395AATGGAATCGAATGGAATCATCATCAAATGGAATCTA7009
ATGGAATCATTGAACGGAATTAAATGGAATCGTCATC
GAATGA
ATTCAATGCAATCAACGAATGGTCTCGAATGGAACCAC
TEE-396AATTGCAAAAGAAACACACATATACACATATAAAACT7010
CAAGAAAGACAAAACTAACCTATGGTGATAGAAATCA
GAAAA
GTACAGTACATTGGTTGTCTTGGTGGG
TEE-397TGACATCATTATTATCAAGAAACATTCTTACCACTGTT7011
ACCAACTTCCCAACACAGACTATGGAGAGAGAGATAA
GACAGA
ATAGCATT
TEE-398AAAGAATTGAATTGAATAGAATCACCAATGAATTGAA7012
TCGAATGGAATCGTCATCGAATGGAATCGAAGGGAAT
CATTGG
ATGGGCTCA
TEE-399ATCATCGAATGGAATCGAATGGAATCAATATCAAACG7013
GAAAAAAACGGAATTATCGAATGGAATCGAATAGAAT
CATCGA
ATGGACC
TEE-400GAATGAAATCGTATAGAATCATCGAATGCAACTGAAT7014
GGAATCATTAAATGGACTTGAAAGGAATTATTATGGA
ATGGAA
TTG
TEE-401TAAGCAACTTCAGCAAAGTCTCAGGATACAAAATCAA7015
TGTGCAAAAATCTCAAGCATTCTTATACACGAACAACA
GACAA
ACAGAGAGCT
TEE-402ACTCAAAAGGAATTGATTCGAATGGAATAGAATGGCA7016
AGGAATAGTATTGAATTGAATGGAATGGAATGGACCC
AAATG
TEE-403GAATGGAATTTAAAGGAATAGAATGGAAGGAATCGGA7017
TGGAATGGAATGGAATAGAATGGAGTCGAATGGAATA
GAATC
GAATGGAATGGCATTG
TEE-404TGAGAAAATGATGGAAAAGAGGAATAAAACGAAACA7018
AAACCACAGGAACACAGGTGCATGTGAATGTGCACAG
ACAAA
GATACAGGGCGGACTGGGAAGGAAGTTTCTGCACCAG
AATTTGGGG
TEE-405AACAAAAAATGAGTCAAGCCTTAAATAAAATCAGAGC7019
CAAAAAAGAAGACATTACATCTGATAAGACAAAAATT
CAAAG
GACCATC
TEE-406AACCCAGTGGAATTGAATTGAATGGAATTGAATGGAA7020
TGGAAAGAATCAATCCGAGTCGAATGGAATGGTATGG
AATGGA
ATGGCATGGAATCAAC
TEE-407ATCAACATCAAACGGAAAAAAAACGGAATTATCGAAT7021
GGAATCGAAGAGAATCATCGAATGGACC
TEE-408AAGGAATGGAATGGTACGGAATAGAATGGAATGGAAC7022
GAATTGTAATGGAATGGAATTTAATGGAACGGAATGG
AATGG
AATGGAATCAACG
TEE-409AACGGAATGGAAAGCAATTTAATCAAATGCAATACAG7023
TGGAATTGAAGGGAATGGAATGGAATGGC
TEE-410AATCGAATGGAACGGAATAGAATAGACTCGAATGTAA7024
TGGATTGCTATGTAATTGATTCGAATGGAATGGAATCG
AATGG
AATGCAATCCAATGGAATGGAATGCAATGCAATGGAA
TGGAATCGAACGGAATGCAGTGGAAGGGAATGG
TEE-411TAGCAACATTTTAGTAACATGATAGAAACAAAACAGC7025
AACATAGCAATGCAATAGTAACACAACAGCAACATCA
TAACAT
GGCAGCA
TEE-412AATGGAATCGAAGAGAATGGAAACAAATGGAATGGA7026
ATTGAATGGAATGGAATTGAATGGAATGGGAAGGAAT
GGAGTG
TEE-413AGCAAACAAGTGAATAAACAAGCAAACAAGTGAACA7027
AGCAAACAAGTGAATAAACAAGCAAACAAGTGAACA
AGCAAA
CAAGTGAATAAACAAGCAAACAAGTGAACAAGGAAA
CAAGTGAATAAACAAAGGCTCT
TEE-414AATGGAATCAACACGAGTGCAATTGAATGGAATCGAA7028
TGGAATGGAATGGAATGGAATGAATTCAACCCGAATG
GAATG
GAAAGGAATGGAATC
TEE-415GAATCGAATGGAATCAACATCAAACGGAAAAAAACGG7029
AATTATCGAATGGAATCGAAGAGAATCATCGAATGGA
CC
TEE-416AACACGAATGTAATGCAATCCAATAGAATGGAATCGA7030
ATGGCATGGAATATAAAGAAATGGAATCGAAGAGAAT
GGAAA
CAAACGGAATGGAATTGAATGGAATGGAATTGAATGG
AATGGGAACGAATGGAGTGAAATTG
TEE-417GAATGGAACGGAATAGAACAGACTCGAATGTAATGGA7031
TTGCTATGTAATTGATTCGAATGGAATGGAATCGAATG
GAATG
CAATCCAATGGAATGGAATGCAATGCAATGGAATGGA
ATCGAATGGAATGCAGTGGAAGGGAATGG
TEE-418GAATCGAATGGAATCAATATCAAACGGAAAAAAACGG7032
AATTATCGAATGGAATCGAAGAGAATCATCGAATGGA
CC
TEE-419ATAAACATCAAACGGAATCAAACGGAATTATCGAATG7033
GAATCGAAGAGAATAATCGAATGGACTCAAATGGAGT
CATCTA
ATGGAATGGTATGGAAGAATCCATGGACTCCAACGCA
ATCATCAGCGAATGGAATC
TEE-420AAAAGAAAAGACAAAAGACACCAATTGCCAATACTGA7034
AATGAAAAAACAGGTAATAACTATTGATCCCATGGAC
ATTAA
AATGATGTTGAAGGAACACCAC
TEE-421AATGTCAAGTGGAATCGAGTGGAATCATCGAAAGAAA7035
TCGAATGGAATCGAAGGGAATCATTGGATGGGCTCAA
AT
TEE-422ATCATCGAATGGAATAGAATGGTATCAACATCAAACG7036
GAGAAAAACGGAATTATCGAATGGAATCGAAGAGAAT
CTTCGA
ACGGACC
TEE-423GAATGGAATCATCGCATAGAATCGGATGGAATTATCA7037
TCGAATGGAATCGAATGGTATCAACATCAAACGGAAA
AAAACG
GAATTATCGAATGGAATCGAATTGAATCATCGAACGG
ACCCG
TEE-424AATGGACTCGAATGGAATAATCATTGAACGGAATCGA7038
ATGGAATCATCATCGGATGGAAATGAATGGAATAATC
CATGGA
CTCGAATGCAATCATCATCGAATGGAATCGAATGGAA
TCATCGAATGGACTCG
TEE-425AATGCAATCATCAACTGGCTTCGAATGGAATCATCAAG7039
AATGGAATCGAATGGAATCATCGAATGGACTC
TEE-426AAGAGACCAATAAGGAATAAGTAAGCAACAAGAGGA7040
AGGAGAAAAGGGCAAGAGAGATGACCAGAGTT
TEE-427TGGAATCATCATAAAATGGAATCGAATGGAATCAACA7041
TCAAATGGAATCAAATGGAATCATTGAACGGAATTGA
ATGGAA
TCGTCAT
TEE-428GGAATCATCGCATAGAATCGAATGGAATTATCATCGA7042
ATGGAATCGAATGGAATCAACATCAAACGAAAAAAAA
CCGGA
ATTATCGAATGGAATCGAAGAGAATCATCGAACGGACC
TEE-429AAATCATCATCGAATGGGATCGAATGGTATCCTTGAAT7043
GGAATCGAATGGAATCATCATCAGATGGAAATGAATG
GAATC
GTCAT
TEE-430GGAATGTAATAGAACGGAAAGCAATGGAATGGAACGC7044
ACTGGATTCGAGTGCAATGGAATCTATTGGAATGGAAT
CGAAT
GGAATGGTTTGGCATGGAATGGAC
TEE-431AAACAATGGAAGATAATGGAAAGATATCGAATGGAAT7045
AGAATGGAATGGAATGGACTCAAATGGAATGGACTTT
AATGG
AATGG
TEE-432GGAACGAAATCGAATGGAACGGAATAGAATAGACTCG7046
AATGTAATGGATTGCTATGTAATTGATTCGAATGGAAT
GGAAT
CGAATGGAATGCAATCCAATGGAATGGAATGCAATGC
AATGAATGGAATGGAATGGAATGGAATGGAA
TEE-433AAACCGAATGGAATGGAATGGACGCAAAATGAATGGA7047
ATGGAAGTCAATGGACTCGAAATGAATGGAATGGAAT
GGAAT
GGAATG
TEE-434GGAATCGAATGGAATCAACATCAAACGGAAAAAAACA7048
GAATTATCGTATGGAATCGAATAGAATCATCGAATGG
ACC
TEE-435CAACCCGAGTGGAATAAAATGGAATGGAATGGAATGA7049
AATGGAATGGATCGGAATGGAATCCAATGGAATCAAC
TGGAA
TGGAATGGAATGGAATG
TEE-436TATCATCGAATGGAATCGAATGGAATCAACATCAAAC7050
GGAAAAAAACGGAATTATCGAATGGAATCGAAGAGAA
TCATC
GAATGGACC
TEE-437CGGAATAATCATTGAACGGAATCGAATGGAATCATCA7051
TCGGATGGAAACGAATGGAATCATCATCGAATGGAAA
TGAAAG
GAGTCATC
TEE-438CAACACACAGAGATTAAAACAAACAAACAAACAATCC7052
AGCCCTGACATTTATGAGTTTACAGACTGGTGGAGAGG
CAGAG
AAG
TEE-439CACTACAAACCACGCTCAAGGCAATAAAAGAACACAA7053
ACAAATGGAAAAACATTCCATGCTCATGGATGGG
TEE-440AATCGAATGGAATTAACATCAAACGGAAAAAAACGGA7054
ATTATCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-441TGGAAAAGAATCAAATTGAATGGCATCGAACGGAATG7055
GGATGGAATGGAATAGACCCAGATGTAATGGACTCGA
ATGGA
ATG
TEE-442GACTAATATTCAGAATATACAAGGAACTCAAACAACT7056
CAACAGTAGAAAAAAAAACCTGAATAGACATTTCTCA
AAAGAA
GACATACAAATGGCC
TEE-443GGTCCATTCGATGATTCTCTTCGATTCCATTCGATAATT7057
CCGTTTTTTCCCGTTTGATGTTGATTCC
TEE-444GGAACGAAATCGAATGGAACGGAATAGAATAGACTCG7058
AATGTAATGGATTGCTATGTAATTGATTCGAATGGAAT
GGAAT
CGAATGGAATGCAATCCAATGGAATGGAATGCAATGC
AATGAATGGAATGGAATGGAATGGAATGGA
TEE-445AGCAACTTCAGTAAAGTGTCAGGATACAAAATCAATG7059
TGCAAAAATCACAAGCATTCTTATACATCAATAACAGA
CAAAC
AGAGAGCCAAA
TEE-446GAATAATCATTGAACGGAATCGAATGGAATCATCATC7060
GGATGGAAACGAATGGAATCATCATCGAATGGAAATG
AAAGG
AGTCATC
TEE-447TAATCATCTTCGAATTGAAAACAAAGCAATCATTAAAT7061
GTACTCTAACGGAATCATCGAATGGACC
TEE-448GGAATCGAATGGAATCAACATCAAACGGAAAAAAACG7062
GAATTATCGAATGGAATCGAAGAGAATCATCGAATGG
ACC
TEE-449AGAGAAAAGATGATCATGTAACCATTGAAAAGACAAT7063
GTACAAAACTAATACTAATCACACAGGACCAGAAAGC
AATTTA
GACCAT
TEE-450AATGGAATCGAATGGAATCAACATCAAACGGAAAAAA7064
CGGAATTATCGAATGGAATCAAAGAGAATCATCGAAT
GGACC
TEE-451AATGGAATTATCATCGAATGGAATCGAATGGAATCAA7065
CATCAAACGGAAAAAAACGGAATTATCGAATGGAATC
GAAGA
GAATCATCGAATGGACC
TEE-452GTCAACACAGGACCAACATAGGACCAACACAGGGTCA7066
ACACAGGACCAACATAGGACCAACACAGGGTCAACAC
AAGAC
CAACATGGGACCAACACAGGGTCAACATAGGACCAAC
ATGGGACCAACACAGGGTCAACACAGGACCAAC
TEE-453GAATCAACTCGATTGCAATCGAATGGAATGGAATGGT7067
ATTAACAGAATAGAATGGAATGGAATGGAATGGAACG
GAACG
TEE-454ACTCGAATGCAATCAACATCAAACGGAATCAAACGGA7068
ATTATCGAATGGAATCGAAGAGAATCATCGAACGGAC
TCGAAT
GGAATCATCTAATGGAATGGAATGG
TEE-455AATGGAATGGAATAATCGACGGACCCGAATGCAATCA7069
TCATCGTACAGAATCGAATGGAATCATCGAATGGACT
GGAATG
GAATGG
TEE-456AATACAAACCACTGCTCAACGAAATAAAAGAGGATAC7070
AAACAAATGGAAGAACATTCTATGCTCATGGGTAGGA
TGAATT
CATATCGTGAAAATGGCCATACTGCC
TEE-457AAACACGCAAACACACACACAAGCACACTACCACACA7071
AGCGGACACACATGCAAACACGCGAACACACACACAT
ATACA
CACAAGCACATTACAAAACACAAGCAAACACCAGCAG
ACACACAAACACACAAACATACATGG
TEE-458AATCGAACGGAATCAACATCAAACGGAAAAAAAACGG7072
AATTATCGAATGGAATCGAAGAGAATCATCGAATGGA
CC
TEE-459TAATTGATTCGAATGGAATGGAATAGAATGGAATTGA7073
ATGGAATGGACCATAATGGATTGGACTTTAATAGAAA
GGGCATG
TEE-460AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG7074
TACAAAAGTCACAAGCATTCTTATACACCAACAAAAG
ACAAAC
AGAGAGCC
TEE-461ACATCAAACGGAAAAAAAAAACAAAACGGAATTATCG7075
AATGGAATCGAAGAGAATCATCGAATGGACC
TEE-462GAAATTCCAATTAAAATGAAATCGACTTATCTTAACAA7076
ATATAGCAATGCTGACAACACTTCTCCGGATATGGGTA
CTGCT
TEE-463ACATCTCACTTTTAGTAATGAACAGATCATTCAGACAG7077
AAAATTAGCAAAGAAACATCAGAGTTAAACTACACTC
TAAAC
CAAATGGACCTA
TEE-464GAAGAAAGCATTCATTCAAGACATCTAACTCGTTGATA7078
TAATGCATACAGTTCAAAATGATTACACTATCATTACA
TCTAG
GGCTTTC
TEE-465ACACACACATTCAAAGCAGCAATATTTACAACAGCCA7079
AAAGGTGGAAACAATTGAGCAATTG
TEE-466ATCATCGAATAGAATCGAATGGTATCAACACCAAACG7080
GAAAAAAACGGAATTATCGAATGGAATCGAAGAGAAT
CTTCGA
ACGGACC
TEE-467ATCAACATCAAACGGAAAAAACGGAATTATCGAATGG7081
AATCGAAGAGAATCATCGAACGGACC
TEE-468AATCGAAAGGAATGTCATCGAATGGAATGGACTCAAA7082
TGGAATAGAATCGGATGGAATGGCATCGAATGGAATG
GAATG
GAATTGGATGGAC
TEE-469AACATGAACAGTGGAACAATCAGTGAACCAATACAAG7083
GGTTAAATAAGCTAGCAATTAAAAGCTGTATCACTGGT
CTAAA
GATAGAAGATCAAGTAGAAAATCAGCGCAAGAGGAA
AGATATACGAAAACTAATGGCC
TEE-470CGAATGGAATCATTATGGAATGGAATGAAATGGAATA7084
ATCAAATGGAATTGAATGGAATCATCGAATGGAATCG
AACAAA
ATCCTCTTTGAATGGAATAAGATGGAATCACCAAATGG
AATTG
TEE-471AAGGGAATTGAATAGAATGAATCCGAATGGAATGGAA7085
TGGAATGGAATGGAATGGAATGGAATGGAATGGAATG
GAATG
TEE-472GAATGGAATCGAATCAAATTAAATCAAATGGAATGCA7086
ATAGAAGGGAATACAATGGAATAGAATGGAATGGAAT
GGAAT
GGACT
TEE-473AAACGGAATCAAACGGAATTATCGAATGGAATCGAAG7087
AGAATCATCGAACGGACTCGAATGGAATCATCTAATG
GAATG
GAATGGAAGAATCCATGGACT
TEE-474ATGGAATCAACATCAAACGGAAAAAAAAACGGAATTA7088
TCGAATGGAATCGAAGAGAATCATCGAATGGACCAGA
ATGGA
ATCATCTAATGGAATGGAATGG
TEE-475AATGGAATCATCATCGAATGGAATCGAATGGAATCAT7089
GGAATGGAATCAAATGGAATCAAATGGAATCGAATGG
AATGG
AATGGAATG
TEE-476AACGGAATCAAACGGAATTACCGAATGGAATCGAATA7090
GAATCATCGAACGGACTCGAATGGAATCATCTAATGG
AATGGA
ATGGAAG
TEE-477AAACGGAATCAAACGGAATTATCGAATGGAATCGAAA7091
AGAATCATCGAACGGACTCGAATGGAATCATCTAATG
GAATG
GAATGGAAGAATCCATGG
TEE-478GAATGATACGGAATACAATGGAATGGAACGAAATGAA7092
ATGGAATGGAATGGAATGGAATGGAATGGAATGG
TEE-479ACAGCAAGAGAGAAATAAAACGACAAGAAAACTACA7093
AAATGCCTATCAATAGTTACTTTAAATATCAGTGGACC
AAATCA
GTGAAACAAAAGACACAGAGTGGC
TEE-480AATGGACTCGAATGGATTAATCATTGAACGGAATCGA7094
ATGGAATCATCATCGGATGGTAATGAATGGAATCATC
ATCGAA
TGGAATCGG
TEE-481GAATGGAATCGAAAGGAATGTCATCGAATGGAATGGA7095
ATGGAACGGAATGGAATCGAATGGAATGGACTCGAAT
GGAAT
AGAATCGAATGCAATGGCATCG
TEE-482ATCGAATGGAATCAACATCAGACGGAAAAAAACGGAA7096
TTATCAAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-483AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG7097
TGCAAAAATCAAAAGCATTCTTATGCACCAATAACAG
ACACAG
AGCCAAAT
TEE-484AATGGAATGGAACGCAATTGAATGGAATGGAATGGAA7098
CGGAATCAACCTGAGTCAAATGGAATGGAATGGAATG
GAATG
TEE-485GGAACGAAATCGAATGGAACGGAATAGAATAGACTCG7099
AATGTCATGGATTGCTATGTAATTGATTGGAATGGAAT
GGAAT
CG
TEE-486TAGCAGGAAACAGCAAACTCAAATTAAGTAATTTCAA7100
GAGCGTATCATCAATGAACTATTTTCAAAGATGTGGGC
AAGAT
TEE-487GAATTGAAAGGAATGTATTGGAATAAAATGGAATCGA7101
ATAGGTTGAAATACCATAGGTTCGAATTGAATGGAAT
GGGAGG
GACACCAATGGAATTG
TEE-488AAGCAACTTCAGCAAAGTCTCGGGATACAAAATCAAT7102
GTGCAAAAATCACAAGCATTCTTATACACCACTAACAG
ACAAA
TGGAGAGTC
TEE-489GAATGGAATCAACATCAAACGGAAAAAAACGGAATTA7103
TCGAATGGAATCGAAGAGAATCATCGAATGGACCAGA
ATGGA
ATCATCTAATGGAATGGAATGGAATAATCCATGG
TEE-490AAAAGCAATTGGACTGATTTTAAATATACGTGGCAAC7104
AAGGATAAACTGCTAATGATGGGTTTGCAAATACAGA
TCG
TEE-491AATGGAATCAACATCGAACGGAAAAAAACGGAATTAT7105
CGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-492AAACGGAATTATCAAATGGAATCGAAGAGAATCATCG7106
AACGGACTCGAATGGAATCATCTAATGGAATGGAATG
GAAG
TEE-493TGCAAGATAACACATTTTAGTTGACACCATTGAAAACA7107
GTTTTAACCAAGAATATTAGAACCAATGAAGCAGAGA
AATCA
AAAGGGTGGATGGAACTGCCAAAGGATG
TEE-494TAGAACAGAATTGAATGGAATGGCATCAAATGGAATG7108
GAAACGAAAGGAATGGAATTGAATGGACTCAAATGTT
ATGGA
ATCAAAGGGAATGGACTC
TEE-495AAGAGAATCATCGAATGGAATCGAATGGAATCAACAT7109
CAAACGGAAAAAAACGGAATTATCGAATGGAATCGAA
GAGAA
TCATCGAATGGACC
TEE-496ATCAACATCAAACGGAAAAAAACGGAATTATCGAATG7110
GAATCGAAGAGAATCATCGAATGGACC
TEE-497GAATCAACATCAAACGGAAAAAAACCGAATTATCGAA7111
TGGAATCGAAGAGAATCATCGAATGGACC
TEE-498ATCAACATCAAACGGAATCAAACGGAATTATCGAATG7112
GAATCGAAGAGAATCATCAAATGGACTCGAATGGAAT
CATCTA
ATGGAATGGAATGGAAGAATCCATGG
TEE-499ATCGAATGGAATCATTGAATGGAAAGGAATGGAATCA7113
TCATGGAATGGAAACGAATGGAATCACTGAATGGACT
CGAATG
GGATCATCA
TEE-500ATTCAGCCTTTAAAAAAAGAAGACAGTCCTGTCATTTG7114
TGACAATATGAATGAAACAGACATCACATTAAATGAA
ATGAG
CCAGGCGCAG
TEE-501GAATGAAATGAAATCAAATGGAATGTACATGAATGGA7115
ATAGAAAAGAATGCATCTTTCTCGAACGGAAGTGCATT
GAATG
GAAAGGAATCTACTGGAATGGATTCGAATGGAATGGA
ATGGGATGGAATGGTATGG
TEE-502AACATCAAACGGAATCAAACGGAATTATCGAATGGAA7116
TCGAAGAGAATCATCGAACGGACTCGAATGGAATCAT
CTAATG
GAATGGAATGGAAGAATCCATGGACTCGAATGCAATC
ATCATCGAATGAAATCGAATGGAATCATCGAATGGAC
TCG
TEE-503ATGGAATTCAATGGAATGGACATGAATGGAATGGACT7117
TCAATGGAATGGTATCAAATGGAATGGAATTCAGT
TEE-504AATGGAAAGGAATCGAATGGAAGGGAATGAAATTGAA7118
TCAACAGGAATGGAAGGGAATAGAATAGACGGCAATG
GAAT
GGACTCG
TEE-505AGCAACTTCAGCAAAGTATCAGGATACAAAATCAATG7119
TACAAAAATCCCAAGCATTCTTATACACCAACAACAG
ACAAAC
AGAGAGCC
TEE-506AGCAACTTCAGCAAAGTCTCAGGATACAAAATCGATG7120
TGCAAAAATCACAAGCATTCTTATACACCAACAACAG
ATAAAC
AGAGAGCC
TEE-507AACGGAAAAAAAACGGAATTATCGAATGGAATCGAAG7121
AGAATCATCGAATGGACCAGAATGGAATCATCTAATG
GAATG
GAATGGAATAATCCATGGACTCGAATG
TEE-508GGAATCAAACGGAATTATCGAATGGAATCGAAGAGAA7122
TCATAGAACGGACTCAAATGGAATCATCTAATGGAAT
GGAAT
GGGAGAATCCATGGACTCGAATG
TEE-509AATGGAATCAATATCAAACGGAAAAAAACGGAATTAT7123
CGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-510AACGGAATCAAACGGAATTATCGAATGGAATCGAAAA7124
GAATCATCGAACGGACTCGAATGGAATCATCTAATGG
AATGG
AATGGAAGAATCCATGG
TEE-511AAACGGAATTATCGAATGGAATCAAAGAGAATCATCG7125
AATGGCCACGAATGGAATCATATAATGGAATGGAATG
GAATA
ATCCATGGACC
TEE-512AATGGAATCGAATGGATTGATATCAAATGGAATGGAA7126
TGGAAGGGAATGGAATGGAATGGAATTGAACCAAATG
TAATG
GATTTG
TEE-513TAAAAGACGGAACAGATAGAAAGCAGAAAGGAAAGG7127
TGAATTGCATTACCACTATTCATACTGCCACACACATG
ACATTA
GGCCAAGTC
TEE-514AATGGAATCGAATGGAACAATCAAATGGACTCCAATG7128
GAGTCATCTAATGGAATCGAGTGGAATCATCGAATGG
ACTCG
TEE-515TAACACATAAACAAACACAGAGACAAAATCTCCGAGA7129
TGTTAATCTGCTCCAGCAATACAGAACAATTTCTATTA
CCAAC
AGAATGCTTAATTTTTCTGCCT
TEE-516GGAATCGAATGGAATCAACATCAAACGGAAAAAAACG7130
GAATTATCGAATGGAATCAAAGAGAATCATCGAATGG
ACC
TEE-517AGAATGGAAAGGAATCGAAACGAAAGGAATGGAGAC7131
AGATGGAATGGAATG
TEE-518GAATCATCATAAAATGGAATCGAATGGAATCAACATC7132
AAATGGAATCAAATGGTCTCGAATGGAATCATCTTCAA
ATGGA
ATGGAATGG
TEE-519AACAACAATGACAAACAAACAACAACGACAAAGACAT7133
TTATTTGGTTCACAAATCTCCAGGGTGTACAAGAAGCA
TGGTG
CCAGCATCTGCTCAGCTTCTGATGAGGGCTCTGGGAAG
CTTTTACTC
TEE-520AACGGACTCGAACGGAATATAATGGAATGGAATGGAT7134
TCGAAAGGAATGGAATGGAATGGACAGGAAAAGAATT
GAATG
GGATTGGAATGGAATCG
TEE-521AACATCAAACGAAATCAAACGGAATTATCAAATTGAA7135
TCGAAGAGAATCATCGAATTGCCACGAATGCAATCAT
CTAATG
GTATGGAATGGAATAATCCATGGACCCAGATG
TEE-522AGAAATTAACAGCAAAAGAAGGATGCAGTGCAACTCA7136
GGACAACACATACAATTCAAGCAACAAATGTATAGTG
GCTGG
GCACCAAGGATACAG
TEE-523GCAATAAAATCGACTCAGATAGAGAAGAATGCAATGG7137
AATGGAATGGAATGGAATGGAATGGGATGGAATGGTA
TGGAA
TGG
TEE-524AATGGACTCGAATGAAATCATCATCAAACGGAATCGA7138
ATGGAATCATTGAATGGAAAGGATGGGATCATCATGG
AATGGA
AACGAATGGAATCACTG
TEE-525CCACATAAAACAAAACTACAAGACAATGATAAAGTTC7139
ACAACATTAACACAATCAGTAATGGAAAAGCCTAGTC
AATGGC
AG
TEE-526TGGAATGGAATGGAATGGAATCAAATCGCATGGTAAT7140
GAATCAAATGGAATCAAATCGAATGGAAATAATGGAA
TCGAA
GGGAAACGAATGGAATCGAATTGCACTGATTCTACTG
ACTTCGAGGAAAATGAAATGAAATGCGGTGAAGTGGA
ATGG
TEE-527GAATGTTATGAAATCAACTCGAACGGAATGCAATAGA7141
ATGGAATGGAATGGAATGGAATGGAATGGAATGG
TEE-528AATGGAATCATTGAATGGAATGGAATGGAATCATCAA7142
AGAAAGGAATCGAAGGGAATCATCGAATGGAATCAAA
CGGAA
TCATCGAATGGAATGGAATGGAATG
TEE-529GGAATCAACATCAAACGGAAAAAAAACGGAATTATCG7143
AATGGAATCGAAGAGAATCATCGAATGGACC
TEE-530GGAATAATCATCATCAAACAGAACCAAATGGAATCAT7144
TGAATGGAATCAAAGGCAATCATGGTCGAATG
TEE-531GCATAGAATCGAATGGAATTATCATTGAATGGAATCG7145
AATGGAATCAACATCAAACGGAAAAAAACGGAATTAT
CGAATG
GAATCGAAGAGAATCATCGAATGGACCC
TEE-532AATGGAATCGAAGAGAATCATCGAACGGACTCGAATG7146
GAATCATCTAATGGAATGGAATGGAATAATCCATGGA
CCCGAA
TG
TEE-533AAATGAATCGAATGGAATTGAATGGAATCAAATAGAA7147
CAAATGGAATCGAAATGAATCAAATGGAATCGAATCG
AATGG
AATTGAATGGCATGGAATTG
TEE-534AGTTAATCCGAATAGAATGGAATGGAATGCAATGGAA7148
CGGAATGGAACGGAATGGAATGGAATGGAATGGAATG
GAATG
TEE-535ATCACAATCACACAACACATTGCACATGCATAACATGC7149
ACTCACAATACACACACAACACATACACAACACACAT
GCAAT
ACAACACAAAACGCAACACAACATATACACAACACAC
AGCACACACATGCC
TEE-536AAAGACTTAAACGTTAGACCTAAAACCATAAAAACCC7150
TAGAGGAAAACCTAGGCATTACCATTCAGGACTTAGG
CATGGG
CAAGGAC
TEE-537AAAGTCCAAAGATGAACAAAATATCCAGAAGGAAAAC7151
AAATGCACTTGGGGAGTGGGAAAGAAAACCAAGACTG
AGCAA
TGCGTCAAGCTCAGACGTGCCTCACTACG
TEE-538AAACGGAATCAAACGGAATTATCGAATGGAGTCGAAA7152
AGAATCATCGAACGGACTCGAATGGAATCATCTAATG
GAATG
GAATGGAAGAATCCATGG
TEE-539AATTGATTCGAAATTAATGGAATTGAATGGAATGCAAT7153
CAAATGGAATGGAATGTAATGCAATGGAATGTAATAG
AATGG
AAAGCAATGGAATG
TEE-540TACAGAACACATGACTCAACAACAGCAGAAAGCATAT7154
TCTTTTCAAATGCACATGAAACATTATCATGATGGACC
AAAT
TEE-541GGAACAAAATGAAATCGAACGGTAGGAATCATACAGA7155
ACAGAAAGAAATGGAACGGAATGGAATG
TEE-542AACGGAAAAAACGGAATTATCGAATGGAATCGAAGAG7156
AATCATCGAATGGAATCGAATGGAGTCATCG
TEE-543AATCGAACGGAATCAACATCAAACGGAAAAAAACGGA7157
ATTATCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-544AGAATGGAATGCAATAGAATGGAATGCAATGGAATGG7158
AGTCATCCGTAATGGAATGGAAAGGAATGCAATGGAA
TGGAA
TGGAATGG
TEE-545ATGGAATCAACATCAAACGGAATCAAACGGAATTATC7159
GAATGGAATCGAAGAGAATCATCGAACGGATTCGAAT
GGAATC
ATCTAATGGAATGGAATGGAAGAATCCATGGACTCGA
ATGCAATCATCAGCGAATGGAATCGAATGGAATCATC
GAATGG
ACTCG
TEE-546GGAATAAAACGGACTCAATAGTAATGGATTGCAATGT7160
AATTGATTCGATTTCGAATGGAATCGCATGGAATGTAA
TGGAA
TGGAATGGAATGGAAGGC
TEE-547AATGGAATCAACATCAAACGGAAAAAAACGGAATTAT7161
CGTATGGAATCGAAAAGAATTATCGAATGGACC
TEE-548TCAAACGGAAAAAAACGGAATTATCGAATGGAATCGA7162
AGAGAATCATCGAATGGACC
TEE-549ACATCAAACGGAATCAAACGGAATTATCGAATGGAAT7163
CGAAAAGAATCATCGAACGGACTCGAATGGAATCATC
TAATGG
AATGGAATGGAAGAATCCATGGACTCGAATG
TEE-550TGGAATCGAATGGAATCAACATCAAACGGAAAAAAAC7164
GGAATTATCGAATGGAATCGAAGAGAATCATCGAATG
GACC
TEE-551AATGGAATCGAATGCAATCATCGAACGGAATCGAATG7165
GCATCACCGAATGGAATGGAATGGAATGGAATGGAAT
GG
TEE-552AGAATTGATTGAATCCAAGTGGAATTGAATGGAATGG7166
AATGGATTAGAAAGGAATGGAATGGATTGGAATGGAT
TGGAAT
GGAAAGG
TEE-553AACTGCATCAACTAACAGGCAAAATAACCAGCTAATA7167
TCATAATGACAGGATTAAATTCACAAATGACAATATTA
ACCGT
AAATGTAAATGGGCTA
TEE-554GTAAACAAACAATCAAGCAAGTAAGAACAGAAATAAC7168
AGCATTTGGCTTTTGAGTTAATGACAAGAACACTCGGC
ATGGG
AGCCTGGGTGAGCAAATCACAGATCTTC
TEE-555AAAGGAATGGACTGGAACAAAATGAAATCGAACGGTA7169
GGAATCGTACAGAACGGACAGAAATGGAACGGCATGG
AATGC
ACTCG
TEE-556GAATCAACCCGAGCGGAAAGGAATGGAATGGAATGGA7170
ATCAACACGAATGGAATGGAACGGAATGGAATGGGAT
GGGAT
GAAATGGAATGG
TEE-557AAGAAATGGAATCGAAGAGAATGGAAACAAACGGAA7171
TGGAATTGAATGGAATGGAATTGAATGGAATGGGA
TEE-558GACATGCAAACACAACACACAGCACACATGGAACATG7172
CATCAGACATGCAAACACAACACACATACCACACATG
GCATAT
GCATCAGACGTGCCTCACTAC
TEE-559AAAGGAATGCACTCGAATGGAATGGACTTGAATGGAA7173
TGTCTCCGAATGGAACAGACTCGTATGAAATGGAATC
GAATGG
AATGGAATCAAATGGAATTGATTTGAGTGAAATGGAA
TCAAATGGAATGGCAACG
TEE-560GGAACAAAATGAAATCGAACGGTAGGAATCGTACAGA7174
ACGGAAAGAAATGGAACGGAATGGAATGCACTCGAAT
GGAAA
GGAGTCCAAT
TEE-561AAATTGATTGAAATCATCATAAAATGGAATCGAAGGG7175
AATCAACATCAAATGGAATCAAATGGAATCATTGAAC
GGAATT
GAATGGAATCGTCAT
TEE-562AGAATGGAAAGCAATAGAATGGAACGCACTGGATTCG7176
AGTGCAATGGAATCAATTGGAATGGAATCGAATGGAA
TGGAT
TGGCA
TEE-563AACACCAAACGGAAAAAAACGGAATTATCGAATGGAA7177
TCGAAGAGAATCTTCGAACGGACCCGAATGGGATCAT
CTAAT
GGAATGGAATGGAATAATCCATGG
TEE-564AATGGAGACTAATGTAATAGAATCAAATGGAATGGCA7178
TCGAATGGAATGGACTGGAATGGAATGTGCATGAATG
GAATGG
AATCGAATGGATTG
TEE-565AAATCGAATGGAACGCAATAGAATAGACTCGAATGTA7179
ATGGATTGCTATGTAATTGATTCGAATGGAATGGAATC
GACTG
GAATGCAATCCAATGGAATGGAATGCAATGCAATGGA
ATGGAATCGAACGGAATGCAGTGGAAGGGAATGG
TEE-566AATCAACAAGGAACTGAAACAAGTAAACAAGAAAAC7180
AAATAACACCATAAAACATGGGCAAAGGACATAAACA
GACATT
TTTCAAAAAAGACATACAAATGGCCGAG
TEE-567AATGGAATCAACATCAAACGGAAAAAAACGGAATTAT7181
CGAATGGAATCGAAGAGAATCATCGAATGGACCCAGG
CTGGT
CTTGAACTCC
TEE-568ATTGAATGGGCTAGAATGGAATCATCTTTGAACGGAAT7182
CAAAGGGAATCATCATCGAATGGAATCGAATGGAAAT
GTCAA
CG
TEE-569AATGGACTCGAATGGAATCAACATCAAATGGAATCAA7183
GCGGAATTATCGAATGAAATCGAAGAGAATCATCGAA
TGGACT
CGAAAGGAATCATCTAATGGAATGGAATGGAATAATC
CATGGACTCGAATGCAATCATCATCG
TEE-570AAACGGAAAAAAACGGAATTATTGAATGGAATCGAAG7184
AGAATCTTCGAACGGACCCGAATGGAATCATCTAATG
GAATG
GAATGGAATAATCCATGG
TEE-571ACTCGAGTGGAATTGACTGTAACAAAATGGAAAGTAA7185
CGGATTGGAATCGAATGGAACGGAATGGAATGGAATG
GACAT
TEE-572TACAAACTTTAAAAAATGATCAACAGATACACAGTTA7186
GCAAGAAAGAATTGAGGGCAAAGAATATGCCAGACAA
ACTCA
AGAGGAAGATGATGGTAGAGATAGGTCACATTGGAGT
GTCA
TEE-573AAATCAACAACAAACGGAAAAAAAAGGAATTATCGAA7187
TGGAATCAAAGAGAATCATCGAATGGACC
TEE-574AACGGAATCAAACGGAATTATCGAATGGAATCGAAAA7188
GAATCATCGAACGGACTCGAATGGAATCATCTAATGT
AATGGA
ATGGAAGAATCCATGGACTCGAATG
TEE-575AACGGAAAAAAACGGAATTATCGAATGGAATCGAAGA7189
GAATCATCGAATGGACCAGAATGGAATCATCTAATGG
AATGG
AATGGAATAATCCATGGACTCGAATG
TEE-576CAACATCAAACGGAAAAAAACGGAATTATGGAATGGA7190
ATCGAAGAGAATCATCGAATGGACCCGAATGGAATCA
TCTGA
AATATAATAGACTCGAAAGGAATG
TEE-577ATGGAATCGAATGGAATGGACTGGAATGGAATGGATT7191
CGAATGGAATCGAATGGAACAATATGGAATGGTACCA
AATG
TEE-578GAATGGAATCAACATCAAACGGAAAAAAACGGAATTA7192
TCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-579AAATGGACTCGAATGGAATCATCATAGAATGGAATCG7193
AATGCAATGGAATGGAATCTTCCGGAATGGAATGGAA
TGGAATGGAATGGAG
TEE-580GAATCATCATAAAATGGAATCGAATGGAATCAACATC7194
AAATGGAATCAAATGGAATCATTGAACGGAATTGAAT
GGAATCGTCAT
TEE-581ATCGAATGGAATCAACATCAAACGGAAAAAAACGGAA7195
TTATCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-582AGCAACTTCAGCAAAGTCTCAGGATACAAAATCAATG7196
TACAAAAATCACAAGCATTCTTATACACCAATAACAG
ACAAACAGAGAGCCAAAA
TEE-583AGAAACAGAAAACAGTCAAACCAATGGGCAATCCATA7197
TCAGATGCAGTATTATGAACAGAAGTGTAAAGAATGC
ACCAGGCACAATGGC
TEE-584GATTGGAACGAAATCGAATGGAACGGAATAGAATAGA7198
CTCGAATGTAATGGATTGCTATGTAATTGATTCGAATG
GAATGGAATCGAATGGAATGCAATCCAATGGAATGGA
ATGCAATGCAATGGAATGG
TEE-585ATGGAATGGAATAATCAACGTACTCGAATGCAATCAT7199
CATCGTATAGAATCGAATGGAATCATCGAATGGACTC
GAATGGAATAATCATTGAACGGAGTCGAATGGAATCA
TCATCGGATGGAAAC
TEE-586AAAGAAATCGAATGGAATCAGTGTCGAATGGAATGGA7200
ATGGAATCGAAGAATTGAATTGAGTAGAATCGAAGGG
AATCATTGGATGGGCTCAAAT
TEE-587AGAAAAGATAACTCGATTAACAAATGAACAAACACCT7201
GAATACACAAGTCTCAAAAGAAGACATAAAAATGGCC
AAC
TEE-588ATGGAATCAACATCAAACGGAATCACACGGAATTATC7202
GAATGGAATCGAAAAGAATCATCGAACGGACTCGAAT
GGAATCATCTAATGGAATGGAATGGAAG
TEE-589AATGGAATCAACATCAAACGGAATCAAGCGAAATTAT7203
CGAATGGAATCGAAGAGAATCATCGAATGGACTCGAA
TGGAATCATCTAATGGAATGGAATGGGAT
TEE-590AAACACAGTACAAATACTAATTCAAATCAAACTTACTC7204
AAAGTCATAATCAAACATGCCAGACGGGCTGAGGGGC
AGCATTA
TEE-591GGAATCGAGTGGAATCATCGAAAGAAATCGAATGGAA7205
TCATTGTCGAATGGAATGGAATGGAATCAAAGAATGG
AATCGAAGGGAATCATTGGATGGGCT
TEE-592AAAGAAAGACAGAGAACAAACGTAATTCAAGATGACT7206
GTTTACATATCCAAGAACATTAGATGGTCAAAGACTTT
AAGAAGGAATACATTCAAAGGCAAAAAGTCACTTACT
GATTTTGGTGGAGTTTGCCACATGGAC
TEE-593GAAAGGAATCATCATTGAATGCAATCACATGGAATCA7207
TCACAGAATGGAATCGTACGGAATCATCATCGAATGG
AATTGAATGGAATCATCAATTGGACTCGAATGGAATC
ATCAAATGGAATCGATTGGAAGTGTCAAATGGACTCG
TEE-594CAATCAGAGCGGACACAAACAAATTGCATGGGAAGAA7208
TCAATATCGTGAAAATGGCC
TEE-595CAGCGCACCACAGCACACACAGTATACACATGACCCA7209
CAATACACACAACACACAACACATTCACACACCAC
TEE-596GCAAACAGAATTCAACACTACATTAGAACGATCATTC7210
ATCACGACCTAGTAGGATGTTTTTCCTGGGATGCAAGG
ATGGTTCAACAT
TEE-597CAATCAAAACAGCAATGAGATACCATTTTACACCAATC7211
AAAATGGCTACTAAAAAGTCAAAAGCAAATGCC
TEE-598TGGAATAGAATGGAATCAATGTTAAGTGGAATCGAGT7212
GGAATCATCGAAAGAAATCGAATGGAATCATTGTCGA
ATGGTATGGAATGGAATCA
TEE-599AATGGAATGGAATCATCGCATAGAATGGAATGGAATT7213
ATCATCGAATTGAATCGAATGGTATCAACATCAAACG
GAAAAAAACGGAAATATCGAATGGAATCGAAGAGAAT
CATCGAACGGACC
TEE-600GAAAAACAAAACAAAACAAACAAACAAACAATCAAA7214
AAAGTGGTAGCAGAAACCAGAAAGTCCATGTATATAG
CTAATTGGCCTGGTTGT
TEE-601AGACCTTTCTCAGAAGACACACAAATTGCCAACAGGT7215
ATATGAAAAAATGTTCAATATCACTAATCATCAGGGCG
ATGCC
TEE-602CATGGAATCGAATGGAATTATCATCGAATGGAATCGA7216
ATGGTACCAACACCAAACGGAAAAAAACGGAATTATC
GAATGGAATCGAAGAGAATCTTCGAACGGACC
TEE-603AGAGCAGAAACAAATGGAATTGAAATGAAGACAACA7217
ATCAAAAGCATCAATGAAATGAAAAGTTGGGTTTTGG
AAGAGAGAAACAAT
TEE-604ACACAAACACACACACACACACACACACACACACACA7218
CACACACACACACACACACACACACACACATAC
TEE-605AACAAACAAATGAGATGATTTCAGATAGTGATAAACA7219
CTATAACATAATTAATTCGTGCCAATCAGAGCATAACA
GTGGTGTGGTGGCTGTGGAACAGATAGCAGAC
TEE-606AATGGAATCGAGTGGAATGGAAGGCAATGGAATAGAA7220
TGGAATGGAATCGAAAGGAACGGAATGGAATGGAATG
GAATG
TEE-607AGAAATGGAATCGGAGAGAATGGAAACAAATGGAAT7221
GGAATTGAATGGAATGGAATTGAATGGAATGGGAACG
TEE-608AAGAGAACTGCAAAACACTGCTCAAAGAAATCAGAGA7222
TGACAAAAACACATGGAAAAACGTTTCATGCTCATGG
ATTGGAAGACTTA
TEE-609AATCAACACGAATAGAATGGAACGGAATGGAATGGAA7223
TGGAATGGAATGGAATGGAGTGGAATGGAACAGAATG
GAGTGGAAT
TEE-610AACATCAAACGAAATCAAACGGAATTATCAAATTGAA7224
TCGAAGAGAATCATCGAATTGCCACGAATGCAACCAT
CTAATGGTATGGAATGGAATAATCCATGGACCCAGATG
TEE-611CGGAATTATCATCGAATGTAATCGAATGGAATCAACAT7225
CAAACGGAAAAAAACGGAATTATCGAATGGAATCGAA
GAGAATCATCGAATGGACC
TEE-612TGGACACACACGAACACACACCTACACACACGTGGAC7226
ACACACGGACACATGGACACACACGAACACATGGACA
CACACACGGGGACACACACAGACACACACAGAGACAC
ACACGGACACATGG
TEE-613ATCAAACGGAATCAAACGGAATTATCGAATGGAATCG7227
AAGAGAATCATCGAATGGACTCGAATGGAATCATCTA
ATGGAATGGAATGGAAGAATCCATGG
TEE-614AAATGGAATGGAATGCACTTGAAAGGAATAGACTGGA7228
ACAAAATGAAATCGAACGGTAGGAATCATACAGAACA
GAAAGAAATGGAACGGAATGGAATG
TEE-615ACCACACACAAAATACACCACACACCACACACACACC7229
ACACACTATACACACACCACACACCACACAC
TEE-616AAAGAAATAGAAGGGAGTTGAACAGAATCGAATGGA7230
ATCGAATCAAATGGAATCGAATGGCATCAAATGGAAT
CGAATGGAATGTGGTGAAGTGGATTGG
TEE-617GGAATCATCATAAAATGGAATCGAATGGAATCATCAT7231
CAAATGGAATCAAATGGAATCATTGAACGGAATTGAA
TGGAATCGTCAT
TEE-618AAAGATCAATGTACAAAAATCAGCAGCATTTCTATAA7232
ACCAACAATGTCCAGGCTGAGAGAGAAATCAAGAAAA
CAATTC
TEE-619TGGAATGGAATGGAATGAAATAAACACGAATAGAATG7233
GAACGGAATGGAACGGAATGGAATGGAATGGAATGG
AAAG
TEE-620TAATCAGCACAATCAACTGTAGTCACAAAACAAATAG7234
TAACGCAATGATAAAGAAACAGAGAACTAGTTCAAAT
AAACATGATAAGATGGGG
TEE-621AAGCGGAATTATCAAATGGAATCGAAGAGAATGGAAA7235
CAAATGGAATGGAATTGAATGGAATGGAATTGAATGG
AATG
TEE-622AATGGAATCAACATCAAACGGAAAAAAACGGAATTAT7236
CGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-623ACTTGAATCGAATGGAAAGGAATTTAATGAACTTAAA7237
TCGAATGGAATATAATGGTATGGAATGGACTCATGGA
ATGGAATGGAAAGGAATC
TEE-624TGGAATCATCATCGAAAGCAAGCGAATGGAATCATCA7238
AATGGAAACGAATGGAATCATCGAATGGACTCGGATG
GAATTGTTGAATGGACT
TEE-625TGGAATCAACATCAAACGGAAAAAAACGGAATTATCG7239
AATGGAATCGAAGAGAATCATCGAATGGACC
TEE-626TAAGTGAATTGAATAGAATCAATCTGAATGTAATGAA7240
ATGGAATGGAACGGAATGGAATGGAATGGAATGGAAT
GGAATGGAATGG
TEE-627AGGAAAATTTAATCAGCAGGAATAGAAACACACTTGA7241
GAAATCCATGTGGAATGAAAAGAGAATGGCTGAGCAG
CAACAGATTGTCAAAAAGGAAATC
TEE-628AACATCAAACGGAAAAAAAACGGAATTATCGAATGGA7242
ATCGAAGAGAATCATCGAATGGACC
TEE-629TAATTGAGAATAAGCATTCCAGTGGAAAAAAAACTAA7243
ACAATTTGTTGTAAAACATCCTTAAAAGCATCAGAAAG
TTAATACAGCAATGAAGAATTACAGGACCAAATTAAG
AATGGTATGGAAGCCTGTTA
TEE-630TATCATCGAATGGAATCGAATGGAATCAACATCAAAC7244
GGAAAAAAACGGAATTATCGAATTGAATCGAAGAGAA
TCATCGAATGGACC
TEE-631AGCAAAACAAACACAATCTGTCGTTCATGGTACTACG7245
ACATACTGGGAGAGATATTCAAATGATCACACAAAAC
AACATG
TEE-632AAGGATTCGAATGGAATGAAAAAGAATTGAATGGAAT7246
AGAACAGAATGGAATCAAATCGAATGAAATGGAGTGG
AATAGAAAGGAATGGAATG
TEE-633AACGGAATCAAACGGAATTATCGAATGGAATCGAAGA7247
GAATCATCGAACGGACTCGAATGGAATCATCTAATGG
AATGGAATGGAAGAATCCATGGACTCGAATGCAATCA
TCATCGAATGGAATCGAACGGAATCATCGAATGGCC
TEE-634AATCAACTAGATGTCAATGGAATGCAATGGAATAGAA7248
TGGAATGGAATTAACACGAATAGAATGGAATGGAATG
GAATGGAATGG
TEE-635AATGGACTCGAATGGAATAATCATTGAACGGAATCGA7249
ATGGAATCATCATCGGATGGAAATGAATGGAATCATC
ATCGCATGGAATCG
TEE-636GAATGGAATGATACGGAATAGAATGGAATGGAACGAA7250
ATGGAATTGAAAGGAAAGGAATGGAATGGAATGGAAT
GG
TEE-637AATCATCATCGAATGGAATCGAATGGTATCATTGAGTG7251
GAATCGAATGGAATCATCATCAGATGGAAATGAATGG
AATCGTCAT
TEE-638GAATCAAATCAATGGAATCAAATCAAATGGAATGGAA7252
TGGAATTGTATGGAATGGAATGGCATGG
TEE-639TAATGCAGTCCAATAGAATGGAATCGAATGGCATGGA7253
ATATAAAGAAATGGAATCGAAGAGAATGGGAACAAAT
GGAATGGAATTGAGTGGAATGGAATTGAATGGAATGG
GAACGAATGGAGTG
TEE-640AACATCAAACGGAAAAAAACGGAATTATCGAATGGAA7254
TCGAAGAGAATCATCGAATGGACC
TEE-641ATCAAAAGGAACGGAATGGAATGGAATGGAATGGAAT7255
GGAATGGAATGGAATGGAATGAAATCAACCCGAATGG
AATGGATTGGCATAGAGTGGAATGG
TEE-642GCCAACAATCATATGAGAAAAAGCTCAACATCACTGA7256
TCATTTCAGGAATGCAAATCAAAACCACAATGAGATA
CTATCA
TEE-643AATCAAATGGAATGAAATCGAATGGAATTGAATCGAA7257
TGGAATGCAATAGAATGTCTTCAAATGGAATCGAATG
GAAATTGGTGAAGTGGACGGGAGTG
TEE-644TAATGGAATCAACATCAAACGGAAAAAAACGGAATTA7258
TCGAATGCAATCGAAGAGAATCATCGAATGGACC
TEE-645AGCAACTTCAGCAAAGTCTCAGCATACAAAATCAATG7259
TGCAAAAATCACACGCATTCCTATACACCAATAACAG
ACAAACAGAGAGCC
TEE-646GAATCAAATGGAATGGACTGTAATGGAATGGATTCGA7260
ATGGAATCGAATGGAGTGGACTCAAATGGAATG
TEE-647AACAAGTGGACGAAGGATATGAACAGACACTTCTCAA7261
GACATTTATGCAGCCAACAGACACACGAAAAAATGCT
CATCATCACTGGCCATCAG
TEE-648AAACGGAAAAAAACGGAATTATCGAATGGAATCGAAT7262
AGAATCATCGAATGGACC
TEE-649TGGAACCGAACAAAGTCATCACCGAATGGAATTGAAA7263
TGAATCATAATCGAATGGAATCAAATGGCATCTTCGAA
TTGACTCGAATGCAATCATCCACTGGGCTT
TEE-650AACGGAATCACGCGGAATTATCGAATGGAATCGAAGA7264
GAATCATCGAATGGACTCGAATGGAATCATCTAATGG
AATGGAATGG
TEE-651GGAATCAACTCGATTGCAATGGAATGCAATGGAAAGG7265
AATGGAATGCAATTAAAGCGAATAGAATGGAATGGAA
TGGAATGGAACGGAATGGAATG
TEE-652AAAACAAACAACAACGACAAATCATGAGACCAGAGTT7266
AAGAAACAATGAGACCAGGCTGGGTGTGGTG
TEE-653AATCGAAAGGAATGCAATATTATTGAACAGAATCGAA7267
AAGAATGGAATCAAATGGAATGGAACAGAGTGGAATG
GACTGC
TEE-654AAGGAATCGAATGGAAGTGAATGAAATTGAATCAACA7268
GGAATGGAAGGGAATAGAATAGACTGTAATGGAATGG
ACTCG
TEE-655AACCCGAGTGCAATAGAATGGAATCGAATGGAATGGA7269
ATGGAATGGAATGGAATGGAATGGAGTC
TEE-656GAATGGAATTGAAAGGAATGGAATGCAATGGAATGGA7270
ATGGGATGGAATGGAATGCAATGGAATCAACTCGATT
GCAATG
TEE-657GAAAAAAACGGAATTATCGAATTGAATCAAATAGAAT7271
CATCGAACGGACCAAAATGGAATCATCTAATGGAATG
GAATGGAATAATCCATGGACTCTAATG
TEE-658TGGAATCATCTAATGGAATGGAATGGAATAATCCATG7272
GACTCGAATGCAATCATCATAAAATGGAATCGAATGG
AATCAACATCAAATGGAATCAAATGGGATCATTGAAC
GGAATTGAATGGAATCGTCAT
TEE-659GAAAAAAACGGAATTATCGAATTGAATCGAATAGAAT7273
CATCGAACGGACCAGAATGGAATCATCTAATGGAATG
GAATGGAATAATCCATGGACTCGAATG
TEE-660AACCACTGCTTAAGGAAATAAGAGAGAACACAAACAA7274
ATGGAAAAACGTTCCATGCTCATGGATAGGAGAATCA
ATATCGTGAAAATGGCC
TEE-661TATCGAATGGAATGGAAAGGAGTGGAGTAGACTCGAA7275
TAGAATGGACTGGAATGAAATAGATTCGAATGGAATG
GAATGGAATGAAGTGGACTCG
TEE-662GTATCAACATCAAACGGAAAAAAACGGAATTATCGAA7276
TGGAATCATCTAATGGAATGGAATGGAATAATCCATG
GACTCGAATG
TEE-663TAAATGGAGACATCATTGAATACAATTGAATGGAATC7277
ATCACATGGAATCGAATGGAATCATCGTAAATGCAAT
CAAGTGGAATCAT
TEE-664GAATGGAATTGAAAGGTATCAACACCAAACGGAAAAA7278
AAAACGGAATTATCGAATGGAATCGAAGAGAATCATC
GAACGGACC
TEE-665AGCAATTTCAGCAAAGTCTCAGGATACAAAATCAATG7279
TACAAATTCACAAGCATTCTTATGGACCAACAACAG
TEE-666GGAATCGAATGGCATCAACATCAAACGGAAAAAAACG7280
GAATTATCGAATGGAATCGAATGGAATCATC
TEE-667AAACAAAACACAGAAATGCAAAGACAAAACATAAAA7281
CGCAGCCATAAAGGACATATTTTAGATAACTGGGGAA
ATTTGTATGGGCTGTGT
TEE-668AATGGAATCAACATCAAACGGAATCAAACGGAATTAT7282
CGAATGGAATCGAAGAGAATCATCGAACGGACTCGAA
TGGAATCATCTAATGGAATGGAATGGAAG
TEE-669AATCGAATGGAATCAGCATCAAACGGAAAAAAACGGA7283
ATTATCGAATGGAATCGAAGAGAATCATCGAATGGACC
TEE-670AAACGGAATTATAGAATGGACTGGAAGAGAATCATCG7284
AACGGACTAGAATGGAATCATCTAATCGAATGGAATG
GAACAATCCATGGTCTAGCA
TEE-671TGAACAGAGAATTGGACAAAACGCACAAAGTAAAGAA7285
AAAGAATGAAGCAACAAAAGCAGAGATTTATTGAAAA
CAAAAGTACACACCACACAGGGTGGGAGTGG
TEE-672ATCATAACGACAAGAACAAATTCACACACAACAATAT7286
TAACTTCAAATCCAAATGGGTTAAATGCTCCAATTAAA
GGATGCAGACGGGCAAATTGGATA
TEE-673ATCATAACGACAAGAACAAATTCACACACAACAATAT7287
TCACTTCAAATCCAAATGGGTTAAATGCTCCAATTAAA
GGATGCAGACGGGCAAATTGGATA
TEE-674GAATGGAATCGAATGGATTGATATCAACTGGAATGGA7288
ATGGAAGGGAATGGAATGGAATGGAATTGAACCAAAT
GTAATGACTTGAATGGAATG
TEE-675GAATCAACATCAAACGGAAAAAAACGGAATTATCGAA7289
TGGAATCGAAGAGAATCATCGAATGGACC
TEE-676GGAATCAACATCAAACGGAAAAAAACGGAATTATCGA7290
ATGGAATCGAAGAGAATCATCGAATGGACC
TEE-677ATGGAATCAACATCAAACGGAATCAAACGGAATTATC7291
GAATGGAATCAAAGAGAATCATCGAACGGACTCGAAT
GGAATCATCTAATGGAATGGAATGGAAGAATCCATGG
ACTCGAATGCAATCATCATCGAAT
TEE-678GGAATGGAATGGAATGGAGCCGAATGGAATGGAATGT7292
ACTCAAATGGAATGC
TEE-679AAAACACCTAGGAATACAGATAACAAGGGACATTAAC7293
TACCTCTTAAAGAGAACTACAAACCACTGCTCAAGGA
AATGAGAGAGGACACAAACACATGGAAAAACATTCCA
TCCTCATGGATAGGAAGAATCAATATTGTGAAAATGG
CC
TEE-680AACACGACTTTGAGAAGAGTAAGTGATTGTTAATTAA7294
AGCAAGAGAATTATTGATGTATCACAGTCATGAGAAA
TATTGGAAGGAATATGGTCCATAC
TEE-681ACACATATCAAACAAACAAAAGCAATTGACTATCTAG7295
AAATGTCTGGGAAATGGCAAGATATTACA
TEE-682GGAATCATCATATAATGGAATCGAATGGAATCAACAT7296
CAAATGGAATCAAATGGAATCATTGAACGGAATTGAA
TGGAATCGTCAT
TEE-683AATGGAATCAACATCAAACGGAATCAAATGGAATTAT7297
CGAATGGAATCGAAGAGAATCATCGAATTGTCACGAA
TGGAATCATCTAATGGAATGGAATGGAATAATCCATG
GCCCCTATGCAATGGACTCGAATGAAATCATCATCAAA
CAGAATCGAATGGAATCATCTAATGGAATGGAATGGC
ATAATCCATGGACTCGAATG
TEE-684TAAAATGAAACAAATATACAACACGAAGGTTATCACC7298
AGAAATATGCCAAAACTTAAATATGAGAATAAGACAG
TCTCAGGGGCCACAGAG
TEE-685AAAATACAGCGTTATGAAAAGAATGAACACACACACA7299
CACACACACACACAGAAAATGT
TEE-686CAAACAAATAGGTACCAAACAAATAACAACATAAACC7300
TGACAACACACTTATTTACAAGAGACATCCCTTATATG
AAAGGGTACAGAAAAGTCGATGGTAAGATGATGGGGA
AAGGTATACCAACCACTAGCAGAAGG
TEE-687TGGAATCGAATGGAATCAATATCAAACGGAAAAAAAC7301
GGAATTATCGAATGGAATCGAAAAGAATCATCGAATG
GGCCCGAATGGAATCATCT
TEE-688ACAAATGGAATCAACAACGAATGGAATCGAATGGAAA7302
CGCCATCGAAAGGAAACGAATGGAATTATCATGAAAT
TGAAATGGATG
TEE-689AATCAATAAATGTAAACCAGCATATAAACAGAACCAA7303
CGACAAAAACCACATGATTATCTCAATAGATGCAGAA
AAGGCC
TEE-690AAAATAAACGCAAATTAAAATCACAAGATACCAACAC7304
ATTCCCACGGCTAAGTACGAAGAACAAGGGCGAATGG
TCAGAATTAAGCTCAAACCT
TEE-691CAACATCAAACGGAATCAAACGGAATTATCGAATGGA7305
ATCGAAGAGAATCATCGAATGGACTCGAATGGAATCA
TCTAATGGAATGGAATGGAAG
TEE-692ACATCAAACGGAAAAAAACGGAATTATCGAATGGAAT7306
CGAAGAGAATCATCGAATGGACC
TEE-693AATGGACTCGAATAGAATTGACTGGAATGGAATGGAC7307
TCGAATGGAATGGAATGGAATGGAAGGGACTCG
TEE-694AAGAAAGACAGAGAACAAACGTAATTCAAGATGACTG7308
ATTACATATCCAAGAACATTAGATGGTCAAAGACTTTA
AGAAGGAATACATTCAAAGGCAAAACGTCACTTACTG
ATTTTGGTGGAGTTTGCCACATGGAC
TEE-695GAATGGAATCGAATGGAATGAACATCAAACGGAAAAA7309
AACGGAATTATCGAATGGAATCAAAGAGAATCATCGA
ATGGACCCG
TEE-696ATGGACTCGAATGTAATAATCATTGAACGGAATCGAA7310
TGGAATCATCATCGGATGGAAACGAATGGAATCATCA
TCGAATGGAATCGAATGGGATC
TEE-697GAAATGGAATGGAAAGGAATAAAATCAAGTGAAATTG7311
GATGGAATGGATTGGAATGGATTGGAATG
TEE-698AAACGGAAAAAAAACGGAATTATCGAATGGAATCGAA7312
GAGAATCATCGAACGAACCAGAATGGAATCATCTAAT
GGAATGGAATGGAATAATCCATGG
TEE-699ATTAACCCGAATAGAATGGAATGGAATGGAATGGAAC7313
GGAACGGAATGGAATGGAATGGAATGGAATGGAATGG
ATCG
TEE-700AACATCAAACGGAAAAAAACGGAATTATCGTATGGAA7314
TCGAAGAGAATCATCGAATGGACC
TEE-701GAATAGAATTGAATCATCATTGAATGGAATCGAGTAG7315
AATCATTGAAATCGAATGGAATCATCATCGAATGGAA
TTGGGTGGAATC
TEE-702CACCGAATAGAATCGAATGGAACAATCATCGAATGGA7316
CTCAAATGGAATTATCCTCAAATGGAATCGAATGGAAT
TATCG
TEE-703AATGCAATCGAATAGAATCATCGAATAGACTCGAATG7317
GAATCATCGAATGGAATGGAATGGAACAGTC
TEE-704AAATCATCATCGAATGGAATCGAATGGTATCATTGAAT7318
GGAATCGAATGGAATCATCATCAGATGGAAATGAATG
GAATCGTCAT
TEE-705GAATGGAATCGAAAGGAATAGAATGGAATGGATCGTT7319
ATGGAAAGACATCGAATGGAATGGAATTGACTCGAAT
GGAATGGACTGGAATGGAACG

MicroRNA controls gene expression through the translational suppression and/or degradation of target messenger RNA. The expression of G-CSF mRNA and Factor 1× mRNA with human or mouse alpha-globin 3′ untranslated regions (UTRs) were down regulated in human primary hepatocytes using miR-122 sequences in the 3′UTR.

Primary human hepatocytes were seeded at a density of 350000 per well in 500 ul cell culture medium (InVitro GRO medium from Celsis, Chicago, Ill.).

G-CSF mRNA having a human alpha-globin 3′UTR (G-CSF Hs3′UTR; mRNA sequence shown in SEQ ID NO: 7320; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a mouse alpha-globin 3′UTR (G-CSF Mm3′UTR; mRNA sequence shown in SEQ ID NO: 7321; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine. G-CSF mRNA containing a human 3′UTR having a miR-122 sequence in the 3′UTR (G-CSF Hs3′UTR miR-122; mRNA sequence shown in SEQ ID NO: 7322; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), or a miR-122 seed sequence in the 3′UTR (G-CSF Hs3′UTR miR-122 seed; mRNA sequence shown in SEQ ID NO: 7323; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF Hs3′UTR miR-122 seedless; mRNA sequence shown in SEQ ID NO: 7324; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine. G-CSF mRNA containing a mouse 3′UTR having a miR-122 sequence in the 3′UTR (G-CSF Mm3′UTR miR-122; mRNA sequence shown in SEQ ID NO: 7325; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), or a miR-122 seed sequence in the 3′UTR (G-CSF Mm3′UTR miR-122 seed; mRNA sequence shown in SEQ ID NO: 7326; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF Mm3′UTR miR-122 seedless; mRNA sequence shown in SEQ ID NO: 7327; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine.

Factor 1× mRNA having a human alpha-globin 3′UTR (Factor 1× Hs3′UTR; mRNA sequence shown in SEQ ID NO: 7328; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a mouse alpha-globin 3′UTR (Factor IX Mm3′UTR; mRNA sequence shown in SEQ ID NO: 7329; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine. Factor 1× mRNA containing a human 3′UTR having a miR-122 sequence in the 3′UTR (Factor 1× Hs3′UTR miR-122; mRNA sequence shown in SEQ ID NO: 7330; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1), or a miR-122 seed sequence in the 3′UTR (Factor IX Hs3′UTR miR-122 seed; mRNA sequence shown in SEQ ID NO: 7331; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-122 sequence without the seed sequence in the 3′UTR (Factor 1× Hs3′UTR miR-122 seedless; mRNA sequence shown in SEQ ID NO: 7332; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine. Factor 1× mRNA containing a mouse 3′UTR having a miR-122 sequence in the 3′UTR (Factor 1× Mm3′UTR miR-122; mRNA sequence shown in SEQ ID NO: 7333; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), or a miR-122 seed sequence in the 3′UTR (Factor 1× Mm3′UTR miR-122 seed; mRNA sequence shown in SEQ ID NO: 7334; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-122 sequence without the seed sequence in the 3′UTR (Factor 1× Mm3′UTR miR-122 seedless; mRNA sequence shown in SEQ ID NO: 7335; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1) were fully modified with 5-methylcytidine and 1-methylpseudouridine.

Each G-CSF or Factor 1× mRNA sequence was tested at a concentration of 500 ng per well in 24 well plates. 24, 48 and 72 hours after transfection, the expression of protein was measured by ELISA. The protein levels for G-CSF are shown in Table 36 and the protein levels for Factor 1× are shown in Table 37.

TABLE 36
G-CSF Protein Expression
Protein Expression (ng/ml)
Description24 Hours48 Hours72 Hours
G-CSF Hs3′UTR43.918.85.7
G-CSF Hs3′UTR miR-1226.90.70.12
G-CSF Hs3′UTR miR-122 seed48.525.68.2
G-CSF Hs3′UTR miR-122 seedless31.711.73.4
G-CSF Mm3′UTR84.9100.421.3
G-CSF Mm3′UTR miR-12224.03.030.8
G-CSF Mm3′UTR miR-122 seed115.896.419.2
G-CSF Mm3′UTR miR-122 seedless113.192.918.9
TABLE 37
Factor IX Protein Expression
Protein Expression (ng/ml)
Description24 Hours48 Hours72 Hours
G-CSF Hs3′UTR43.918.85.7
G-CSF Hs3′UTR miR-1226.90.70.12
G-CSF Hs3′UTR miR-122 seed48.525.68.2
G-CSF Hs3′UTR miR-122 seedless31.711.73.4
G-CSF Mm3′UTR84.9100.421.3
G-CSF Mm3′UTR miR-12224.03.030.8
G-CSF Mm3′UTR miR-122 seed115.896.419.2
G-CSF Mm3′UTR miR-122 seedless113.192.918.9

HeLa and RAW264 cells were seeded at a density of 17000 and 80000 per well respectively, in 100 ul cell culture medium (DMEM+10% FBS).

G-CSF mRNA (G-CSF; mRNA sequence shown in SEQ ID NO: 6595; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) was fully modified with 5-methylcytidine and 1-methylpseudouridine.

G-CSF mRNA having a miR-142-3p sequence in the 3′UTR (G-CSF miR-142-3p; mRNA sequence shown in SEQ ID NO: 6634; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), or a miR-142-3p seed sequence in the 3′UTR (G-CSF miR-142-3p seed; mRNA sequence shown in SEQ ID NO: 6636; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-142-3p sequence without the seed sequence in the 3′UTR (G-CSF miR-142-3p seedless; mRNA sequence shown in SEQ ID NO: 6638; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine.

G-CSF mRNA having a miR-142-5p sequence in the 3′UTR (G-CSF miR-142-5p; mRNA sequence shown in SEQ ID NO: 6628; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), or a miR-142-5p seed sequence in the 3′UTR (G-CSF miR-142-5p seed; mRNA sequence shown in SEQ ID NO: 6630; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-142-5p sequence without the seed sequence in the 3′UTR (G-CSF miR-142-5p seedless; mRNA sequence shown in SEQ ID NO: 6632; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine.

G-CSF mRNA having a miR-146a sequence in the 3′UTR (G-CSF miR-146a; mRNA sequence shown in SEQ ID NO: 6640; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), or a miR-146a seed sequence in the 3′UTR (G-CSF miR-146a seed; mRNA sequence shown in SEQ ID NO: 6642; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a miR-146a sequence without the seed sequence in the 3′UTR (G-CSF miR-146a seedless; mRNA sequence shown in SEQ ID NO: 6644; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with 5-methylcytidine and 1-methylpseudouridine.

Each G-CSF mRNA sequence was tested at a concentration of 500 ng per well in 24 well plates for each cell type. 24 hours after transfection, the expression of protein was measured by ELISA and the protein levels are shown in Table 38. The G-CSF sequences with a miR-142-3p sequence in the 3′UTR down regulated G-CSF expression in RAW264 cells whereas the G-CSF sequences with a miR-142-5p or miR-146a sequence in the 3′UTR did not down regulate G-CSF expression.

TABLE 38
G-CSF Expression
HeLa CellsRAW264 Cells
Protein ExpressionProtein Expression
Description(ng/ml)(ng/ml)
G-CSF243.5173.7
G-CSF miR-142-3p309.167.6
G-CSF miR-142-3p seed259.8178.1
G-CSF miR-142-3p seedless321.7220.2
G-CSF miR-142-5p291.8223.3
G-CSF miR-142-5p seed261.3233.1
G-CSF miR-142-5p seedless330.2255.1
G-CSF miR-146a272.6125.2
G-CSF miR-146a seed219.4138.3
G-CSF miR-146a seedless217.7132.8

HeLa cells were seeded at a density of 17000 per well in 100 ul cell culture medium (DMEM+10% FBS). G-CSF mRNA having an IRES sequence and Kozak sequence (G-CSF IRES Kozak; mRNA sequence shown in SEQ ID NO: 7336; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), G-CSF mRNA having an IRES sequence but not a Kozak sequence (G-CSF IRES; mRNA sequence shown in SEQ ID NO: 7337; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), G-CSF mRNA without an IRES or Kozak sequence (GCSF no Kozak; mRNA sequence shown in SEQ ID NO: 7338; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or a G-CSF sequence having a Kozak sequence (G-CSF Kozak; mRNA sequence shown in SEQ ID NO: 7339; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) were fully modified with fully modified with 5-methylcytidine and 1-methylpseudouridine and tested at a concentration of 75 ng per well in 24 well plates. 24 hours after transfection, the expression of G-CSF was measured by ELISA, and the results are shown in Table 39.

TABLE 39
G-CSF expression
Protein Expression
Description(ng/ml)
G-CSF IRES Kozak2.01
G-CSF IRES1.64
G-CSF no Kozak795.53
G-CSF Kozak606.28

Modified mRNA encoding G-CSF or mCherry with a human or mouse MALAT1 sequence and their corresponding cDNA sequences are shown below in Table 40. In Table 40, the start codon of each sequence is underlined and the MALAT1 sequences are bolded.

TABLE 40
MALAT1 Constructs
SEQ
ID
SequenceNO:
G-CSFOptimized G-CSF cDNA sequence containing a T7 polymerase site,7340
withkozak sequence, and a Mouse MALAT1 sequence (bold):
MouseTAATACGACTCACTATA
MALAT1GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATA
sequenceTAAGAGCCACC
ATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTA
TGGCCCTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGG
ACAGTCCAAGAAGCGACTCCTCTCGGACCTGCCTCAT
CGTTGCCGCAGTCATTCCTTTTGAAGTGTCTGGAGCAG
GTGCGAAAGATTCAGGGCGATGGAGCCGCACTCCAAG
AGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGA
GGAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCC
TGGGCTCCTCTCTCGTCCTGTCCGTCGCAGGCTTTGCA
GTTGGCAGGGTGCCTTTCCCAGCTCCACTCCGGTTTGT
TCTTGTATCAGGGACTGCTGCAAGCCCTTGAGGGAAT
CTCGCCAGAATTGGGCCCGACGCTGGACACGTTGCAG
CTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGC
AGATGGAGGAACTGGGGATGGCACCCGCGCTGCAGCC
CACGCAGGGGGCAATGCCGGCCTTTGCGTCCGCGTTT
CAGCGCAGGGCGGGTGGAGTCCTCGTAGCGAGCCACC
TTCAATCATTTTTGGAAGTCTCGTACCGGGTGCTGAGA
CATCTTGCGCAGCCG
TGATAATAG
GATTCGTCAGTAGGGTTGTAAAGGTTTTTCTTTTCC
TGAGAAAACAACCTTTTGTTTTCTCAGGTTTTGCTT
TTTGGCCTTTCCCTAGCTTTAAAAAAAAAAAAGCAA
AAGTGGTCTTTGAATAAAGTCTGAGTGGGCGGCTCTA
GA
mRNA sequence (transcribed):7341
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAU
AUAAGAGCCACC
AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUU
AUGGCCCUGCAGUUGCUGCUUUGGCACUCGGCCCUC
UGGACAGUCCAAGAAGCGACUCCUCUCGGACCUGCC
UCAUCGUUGCCGCAGUCAUUCCUUUUGAAGUGUCUG
GAGCAGGUGCGAAAGAUUCAGGGCGAUGGAGCCGCA
CUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUGC
CAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUG
GGGAUUCCCUGGGCUCCUCUCUCGUCCUGUCCGUCG
CAGGCUUUGCAGUUGGCAGGGUGCCUUUCCCAGCUC
CACUCCGGUUUGUUCUUGUAUCAGGGACUGCUGCAA
GCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGACG
CUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCA
ACAACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUG
GCACCCGCGCUGCAGCCCACGCAGGGGGCAAUGCCG
GCCUUUGCGUCCGCGUUUCAGCGCAGGGCGGGUGGA
GUCCUCGUAGCGAGCCACCUUCAAUCAUUUUUGGAA
GUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCG
UGAUAAUAG
GAUUCGUCAGUAGGGUUGUAAAGGUUUUUCUUUU
CCUGAGAAAACAACCUUUUGUUUUCUCAGGUUUUG
CUUUUUGGCCUUUCCCUAGCUUUAAAAAAAAAAAA
GCAAAAGUGGUCUUUGAAUAAAGUCUGAGUGGGCG
GC
mCherryOptimized mCherry cDNA sequence containing a T7 polymerase7342
withsite, kozak sequence, and a Mouse MALAT1 sequence (bold):
MouseTAATACGACTCACTATA
MALAT1GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATA
sequenceTAAGAGCCACC
ATGGTATCCAAGGGGGAGGAGGACAACATGGCGATC
ATCAAGGAGTTCATGCGATTCAAGGTGCACATGGAAG
GTTCGGTCAACGGACACGAATTTGAAATCGAAGGAGA
GGGTGAAGGAAGGCCCTATGAAGGGACACAGACCGC
GAAACTCAAGGTCACGAAAGGGGGACCACTTCCTTTC
GCCTGGGACATTCTTTCGCCCCAGTTTATGTACGGGTC
CAAAGCATATGTGAAGCATCCCGCCGATATTCCTGAC
TATCTGAAACTCAGCTTTCCCGAGGGATTCAAGTGGG
AGCGGGTCATGAACTTTGAGGACGGGGGTGTAGTCAC
CGTAACCCAAGACTCAAGCCTCCAAGACGGCGAGTTC
ATCTACAAGGTCAAACTGCGGGGGACTAACTTTCCGT
CGGATGGGCCGGTGATGCAGAAGAAAACGATGGGAT
GGGAAGCGTCATCGGAGAGGATGTACCCAGAAGATG
GTGCATTGAAGGGGGAGATCAAGCAGAGACTGAAGTT
GAAAGATGGGGGACATTATGATGCCGAGGTGAAAAC
GACATACAAAGCGAAAAAGCCGGTGCAGCTTCCCGGA
GCGTATAATGTGAATATCAAGTTGGATATTACTTCACA
CAATGAGGACTACACAATTGTCGAACAGTACGAACGC
GCTGAGGGTAGACACTCGACGGGAGGCATGGACGAG
TTGTACAAA
TGATAATAG
GATTCGTCAGTAGGGTTGTAAAGGTTTTTCTTTTCC
TGAGAAAACAACCTTTTGTTTTCTCAGGTTTTGCTT
TTTGGCCTTTCCCTAGCTTTAAAAAAAAAAAAGCAA
AAGTGGTCTTTGAATAAAGTCTGAGTGGGCGGCTCTA
GA
mRNA sequence (transcribed):7343
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAU
AUAAGAGCCACC
AUGGUAUCCAAGGGGGAGGAGGACAACAUGGCGAUC
AUCAAGGAGUUCAUGCGAUUCAAGGUGCACAUGGAA
GGUUCGGUCAACGGACACGAAUUUGAAAUCGAAGGA
GAGGGUGAAGGAAGGCCCUAUGAAGGGACACAGACC
GCGAAACUCAAGGUCACGAAAGGGGGACCACUUCCU
UUCGCCUGGGACAUUCUUUCGCCCCAGUUUAUGUAC
GGGUCCAAAGCAUAUGUGAAGCAUCCCGCCGAUAUU
CCUGACUAUCUGAAACUCAGCUUUCCCGAGGGAUUC
AAGUGGGAGCGGGUCAUGAACUUUGAGGACGGGGG
UGUAGUCACCGUAACCCAAGACUCAAGCCUCCAAGA
CGGCGAGUUCAUCUACAAGGUCAAACUGCGGGGGAC
UAACUUUCCGUCGGAUGGGCCGGUGAUGCAGAAGAA
AACGAUGGGAUGGGAAGCGUCAUCGGAGAGGAUGU
ACCCAGAAGAUGGUGCAUUGAAGGGGGAGAUCAAGC
AGAGACUGAAGUUGAAAGAUGGGGGACAUUAUGAU
GCCGAGGUGAAAACGACAUACAAAGCGAAAAAGCCG
GUGCAGCUUCCCGGAGCGUAUAAUGUGAAUAUCAAG
UUGGAUAUUACUUCACACAAUGAGGACUACACAAUU
GUCGAACAGUACGAACGCGCUGAGGGUAGACACUCG
ACGGGAGGCAUGGACGAGUUGUACAAA
UGAUAAUAG
GAUUCGUCAGUAGGGUUGUAAAGGUUUUUCUUUU
CCUGAGAAAACAACCUUUUGUUUUCUCAGGUUUUG
CUUUUUGGCCUUUCCCUAGCUUUAAAAAAAAAAAA
GCAAAAGUGGUCUUUGAAUAAAGUCUGAGUGGGCG
GC
G-CSFOptimized G-CSF cDNA sequence containing a T7 polymerase site,7344
withkozak sequence, and a Human MALAT1 sequence (bold):
HumanTAATACGACTCACTATA
MALAT1GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATA
sequenceTAAGAGCCACC
ATGGCCGGTCCCGCGACCCAAAGCCCCATGAAACTTA
TGGCCCTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGG
ACAGTCCAAGAAGCGACTCCTCTCGGACCTGCCTCAT
CGTTGCCGCAGTCATTCCTTTTGAAGTGTCTGGAGCAG
GTGCGAAAGATTCAGGGCGATGGAGCCGCACTCCAAG
AGAAGCTCTGCGCGACATACAAACTTTGCCATCCCGA
GGAGCTCGTACTGCTCGGGCACAGCTTGGGGATTCCC
TGGGCTCCTCTCTCGTCCTGTCCGTCGCAGGCTTTGCA
GTTGGCAGGGTGCCTTTCCCAGCTCCACTCCGGTTTGT
TCTTGTATCAGGGACTGCTGCAAGCCCTTGAGGGAAT
CTCGCCAGAATTGGGCCCGACGCTGGACACGTTGCAG
CTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGC
AGATGGAGGAACTGGGGATGGCACCCGCGCTGCAGCC
CACGCAGGGGGCAATGCCGGCCTTTGCGTCCGCGTTT
CAGCGCAGGGCGGGTGGAGTCCTCGTAGCGAGCCACC
TTCAATCATTTTTGGAAGTCTCGTACCGGGTGCTGAGA
CATCTTGCGCAGCCG
TGATAATAG
TGCTCTTCAGTAGGGTCATGAAGGTTTTTCTTTTCC
TGAGAAAACAACACGTATTGTTTTCTCAGGTTTTGC
TTTTTGGCCTTTTTCTAGCTTAAAAAAAAAAAAAGC
AAAAGTGGTCTTTGAATAAAGTCTGAGTGGGCGGCTC
TAGA
mRNA sequence (transcribed):7345
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAU
AUAAGAGCCACC
AUGGCCGGUCCCGCGACCCAAAGCCCCAUGAAACUU
AUGGCCCUGCAGUUGCUGCUUUGGCACUCGGCCCUC
UGGACAGUCCAAGAAGCGACUCCUCUCGGACCUGCC
UCAUCGUUGCCGCAGUCAUUCCUUUUGAAGUGUCUG
GAGCAGGUGCGAAAGAUUCAGGGCGAUGGAGCCGCA
CUCCAAGAGAAGCUCUGCGCGACAUACAAACUUUGC
CAUCCCGAGGAGCUCGUACUGCUCGGGCACAGCUUG
GGGAUUCCCUGGGCUCCUCUCUCGUCCUGUCCGUCG
CAGGCUUUGCAGUUGGCAGGGUGCCUUUCCCAGCUC
CACUCCGGUUUGUUCUUGUAUCAGGGACUGCUGCAA
GCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGACG
CUGGACACGUUGCAGCUCGACGUGGCGGAUUUCGCA
ACAACCAUCUGGCAGCAGAUGGAGGAACUGGGGAUG
GCACCCGCGCUGCAGCCCACGCAGGGGGCAAUGCCG
GCCUUUGCGUCCGCGUUUCAGCGCAGGGCGGGUGGA
GUCCUCGUAGCGAGCCACCUUCAAUCAUUUUUGGAA
GUCUCGUACCGGGUGCUGAGACAUCUUGCGCAGCCG
UGAUAAUAG
UGCUCUUCAGUAGGGUCAUGAAGGUUUUUCUUUUC
CUGAGAAAACAACACGUAUUGUUUUCUCAGGUUUU
GCUUUUUGGCCUUUUUCUAGCUUAAAAAAAAAAAA
AGCAAAAGUGGUCUUUGAAUAAAGUCUGAGUGGGC
GGC
mCherryOptimized mCherry cDNA sequence containing a T7 polymerase7346
withsite, kozak sequence, and a Human MALAT1 sequence (bold):
HumanTAATACGACTCACTATA
MALAT1GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATA
sequenceTAAGAGCCACC
ATGGTATCCAAGGGGGAGGAGGACAACATGGCGATC
ATCAAGGAGTTCATGCGATTCAAGGTGCACATGGAAG
GTTCGGTCAACGGACACGAATTTGAAATCGAAGGAGA
GGGTGAAGGAAGGCCCTATGAAGGGACACAGACCGC
GAAACTCAAGGTCACGAAAGGGGGACCACTTCCTTTC
GCCTGGGACATTCTTTCGCCCCAGTTTATGTACGGGTC
CAAAGCATATGTGAAGCATCCCGCCGATATTCCTGAC
TATCTGAAACTCAGCTTTCCCGAGGGATTCAAGTGGG
AGCGGGTCATGAACTTTGAGGACGGGGGTGTAGTCAC
CGTAACCCAAGACTCAAGCCTCCAAGACGGCGAGTTC
ATCTACAAGGTCAAACTGCGGGGGACTAACTTTCCGT
CGGATGGGCCGGTGATGCAGAAGAAAACGATGGGAT
GGGAAGCGTCATCGGAGAGGATGTACCCAGAAGATG
GTGCATTGAAGGGGGAGATCAAGCAGAGACTGAAGTT
GAAAGATGGGGGACATTATGATGCCGAGGTGAAAAC
GACATACAAAGCGAAAAAGCCGGTGCAGCTTCCCGGA
GCGTATAATGTGAATATCAAGTTGGATATTACTTCACA
CAATGAGGACTACACAATTGTCGAACAGTACGAACGC
GCTGAGGGTAGACACTCGACGGGAGGCATGGACGAG
TTGTACAAA
TGATAATAG
TGCTCTTCAGTAGGGTCATGAAGGTTTTTCTTTTCC
TGAGAAAACAACACGTATTGTTTTCTCAGGTTTTGC
TTTTTGGCCTTTTTCTAGCTTAAAAAAAAAAAAAGC
AAAAGTGGTCTTTGAATAAAGTCTGAGTGGGCGGCTC
TAGA
mRNA sequence (transcribed):7347
GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAU
AUAAGAGCCACC
AUGGUAUCCAAGGGGGAGGAGGACAACAUGGCGAUC
AUCAAGGAGUUCAUGCGAUUCAAGGUGCACAUGGAA
GGUUCGGUCAACGGACACGAAUUUGAAAUCGAAGGA
GAGGGUGAAGGAAGGCCCUAUGAAGGGACACAGACC
GCGAAACUCAAGGUCACGAAAGGGGGACCACUUCCU
UUCGCCUGGGACAUUCUUUCGCCCCAGUUUAUGUAC
GGGUCCAAAGCAUAUGUGAAGCAUCCCGCCGAUAUU
CCUGACUAUCUGAAACUCAGCUUUCCCGAGGGAUUC
AAGUGGGAGCGGGUCAUGAACUUUGAGGACGGGGG
UGUAGUCACCGUAACCCAAGACUCAAGCCUCCAAGA
CGGCGAGUUCAUCUACAAGGUCAAACUGCGGGGGAC
UAACUUUCCGUCGGAUGGGCCGGUGAUGCAGAAGAA
AACGAUGGGAUGGGAAGCGUCAUCGGAGAGGAUGU
ACCCAGAAGAUGGUGCAUUGAAGGGGGAGAUCAAGC
AGAGACUGAAGUUGAAAGAUGGGGGACAUUAUGAU
GCCGAGGUGAAAACGACAUACAAAGCGAAAAAGCCG
GUGCAGCUUCCCGGAGCGUAUAAUGUGAAUAUCAAG
UUGGAUAUUACUUCACACAAUGAGGACUACACAAUU
GUCGAACAGUACGAACGCGCUGAGGGUAGACACUCG
ACGGGAGGCAUGGACGAGUUGUACAAA
UGAUAAUAG
UGCUCUUCAGUAGGGUCAUGAAGGUUUUUCUUUUC
CUGAGAAAACAACACGUAUUGUUUUCUCAGGUUUU
GCUUUUUGGCCUUUUUCUAGCUUAAAAAAAAAAAA
AGCAAAAGUGGUCUUUGAAUAAAGUCUGAGUGGGC
GGC

These modified mRNA sequences can include at least one chemical modification described herein. The G-CSF or mCherry modified mRNA sequence can be formulated, using methods described herein and/or known in the art, prior to transfection and/or administration.

The modified mRNA sequence encoding G-CSF or mCherry can be transfected in vitro to various cell types such as HEK293, HeLa, PBMC and BJ fibroblast and those described in Table 25 of U.S. Provisional Application No. 61/839,903, filed Jun. 27, 2013, the contents of which are herein incorporated by reference in its entirety, using methods disclosed herein and/or are known in the art. The cells are then analyzed using methods disclosed herein and/or are known in the art to determine the concentration of G-CSF or mCherry and/or the cell viability.

Septin 4 may be an oncology-related polypeptide of interest. Shown in Table 41, in addition to the name and description of the gene encoding the oncology-related polypeptide of interest, are the ENSEMBL Transcript ID (ENST), the ENSEMBL Protein ID (ENSP), each present where applicable, and when available the optimized sequence ID (OPT. SEQ ID).

TABLE 41
Oncology-Related Targets
TargetTrans.Prot.
TargetDescriptionENST IDSEQ ID NOENSP IDSEQ ID NOOPT. SEQ ID NO
SEPT4septin 4317256734832107173557363, 7368, 7375,
7382, 7389, 7396
SEPT4septin 4317268734932167473567364, 7369, 7376,
7383, 7390, 7397,
7403-7489
SEPT4septin 4393086735037680173577370, 7377, 7384,
7391, 7398
SEPT4septin 4412945735141477973587365, 7371, 7378,
7385, 7392, 7399
SEPT4septin 4426861735240234873597366, 7372, 7379,
7386, 7393, 7400
SEPT4septin 4457347735340200073607367, 7373, 7380,
7387, 7394, 7401
SEPT4septin 4583114735446376873617374, 7381, 7388,
7395, 7402
SEPT4septin 47362

Cell lysates containing protein produced from: (a) apoptosis-inducing factor 1, mitochondrial, short isoform (AIFsh; gene name AIFM1) modified mRNA (mRNA sequence shown in SEQ ID NO. 6617 (Table 42); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1); (b) copper metabolism (Murr1) domain containing 1 (COMMD1) modified mRNA (mRNA sequence shown in SEQ ID NO. 7491 (Table 42); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1); (c) septin 4 (SEPT4) modified mRNA (mRNA sequence shown in SEQ ID NO. 7362 (Table 42); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1); and (d) diablo, IAP-binding mitochondrial protein (DIABLO) modified mRNA (mRNA sequence shown in SEQ ID NO. 7494 (Table 42); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1); all fully modified with 5-methylcytidine and pseudouridine (5mC and pU), fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1mpU), modified where 25% of uridine modified with 2-thiouridine and 25% of cytidine modified with 5-methylcytidine (s2U and 5mC), fully modified with pseudouridine (pU), or fully modified with 1-methylpseudouridine (1mpU) were evaluated using the LC-MS/MS with quantitative LC-MRM as described in Example 31.

TABLE 42
Target Sequences
SEQ ID
DescriptionSequenceNO:
AIFshGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAU6617
AAGAGCCACCAUGGAAAAAGUCAGACGAGAGGGGGUU
AAGGUGAUGCCCAAUGCUAUUGUGCAAUCCGUUGGAG
UCAGCAGUGGCAAGUUACUUAUCAAGCUGAAAGACGG
CAGGAAGGUAGAAACUGACCACAUAGUGGCAGCUGUG
GGCCUGGAGCCCAAUGUUGAGUUGGCCAAGACUGGUG
GCCUGGAAAUAGACUCAGAUUUUGGUGGCUUCCGGGU
AAAUGCAGAGCUACAAGCACGCUCUAACAUCUGGGUG
GCAGGAGAUGCUGCAUGCUUCUACGAUAUAAAGUUGG
GAAGGAGGCGGGUAGAGCACCAUGAUCACGCUGUUGU
GAGUGGAAGAUUGGCUGGAGAAAAUAUGACUGGAGCU
GCUAAGCCGUACUGGCAUCAGUCAAUGUUCUGGAGUG
AUUUGGGCCCCGAUGUUGGCUAUGAAGCUAUUGGUCU
UGUGGACAGUAGUUUGCCCACAGUUGGUGUUUUUGCA
AAAGCAACUGCACAAGACAACCCCAAAUCUGCCACAGA
GCAGUCAGGAACUGGUAUCCGAUCAGAGAGUGAGACA
GAGUCCGAGGCCUCAGAAAUUACUAUUCCUCCCAGCAC
CCCGGCAGUUCCACAGGCUCCCGUCCAGGGGGAGGACU
ACGGCAAAGGUGUCAUCUUCUACCUCAGGGACAAAGU
GGUCGUGGGGAUUGUGCUAUGGAACAUCUUUAACCGA
AUGCCAAUAGCAAGGAAGAUCAUUAAGGACGGUGAGC
AGCAUGAAGAUCUCAAUGAAGUAGCCAAACUAUUCAA
CAUUCAUGAAGACUGAUAAUAGGCUGGAGCCUCGGUG
GCCAUGCUUCUUGCCCCUUGGGCCUCCCCCCAGCCCCU
CCUCCCCUUCCUGCACCCGUACCCCCGUGGUCUUUGAA
UAAAGUCUGAGUGGGCGGC
MEKVRREGVKVMPNAIVQSVGVSSGKLLIKLKDGRK7490
VETDHIVAAVGLEPNVELAKTGGLEIDSDFGGFRVNA
ELQARSNIWVAGDAACFYDIKLGRRRVEHHDHAVVS
GRLAGENMTGAAKPYWHQSMFWSDLGPDVGYEAIG
LVDSSLPTVGVFAKATAQDNPKSATEQSGTGIRSESET
ESEASEITIPPSTPAVPQAPVQGEDYGKGVIFYLRDKV
VVGIVLWNIFNRMPIARKIIKDGEQHEDLNEVAKLFNI
HED
COMMD1GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAA7491
UAUAAGAGCCACCAUGGCGGCGGGCGAGCUUGAG
GGUGGCAAACCCCUGAGCGGGCUGCUGAAUGCGC
UGGCCCAGGACACUUUCCACGGGUACCCCGGCAUC
ACAGAGGAGCUGCUACGGAGCCAGCUAUAUCCAG
AGGUGCCACCCGAGGAGUUCCGCCCCUUUCUGGCA
AAGAUGAGGGGGAUUCUUAAGUCUAUUGCGUCUG
CAGACAUGGAUUUCAACCAGCUGGAGGCAUUCUU
GACUGCUCAAACCAAAAAGCAAGGUGGGAUCACA
UCUGACCAAGCUGCUGUCAUUUCCAAAUUCUGGA
AGAGCCACAAGACAAAAAUCCGUGAGAGCCUCAU
GAACCAGAGCCGCUGGAAUAGCGGGCUUCGGGGC
CUGAGCUGGAGAGUUGAUGGCAAGUCUCAGUCAA
GGCACUCAGCUCAAAUACACACACCUGUUGCCAU
UAUAGAGCUGGAAUUAGGCAAAUAUGGACAGGAA
UCUGAAUUUCUGUGUUUGGAAUUUGAUGAGGUCA
AAGUCAACCAAAUUCUGAAGACGCUGUCAGAGGU
AGAAGAAAGUAUCAGCACACUGAUCAGCCAGCCU
AACUGAUAAUAGGCUGGAGCCUCGGUGGCCAUGC
UUCUUGCCCCUUGGGCCUCCCCCCAGCCCCUCCUC
CCCUUCCUGCACCCGUACCCCCGUGGUCUUUGAAU
AAAGUCUGAGUGGGCGGC
MAAGELEGGKPLSGLLNALAQDTFHGYPGITEELLRS7492
QLYPEVPPEEFRPFLAKMRGILKSIASADMDFNQLEAF
LTAQTKKQGGITSDQAAVISKFWKSHKTKIRESLMNQ
SRWNSGLRGLSWRVDGKSQSRHSAQIHTPVAIIELELG
KYGQESEFLCLEFDEVKVNQILKTLSEVEESISTLISQPN
SEPT4GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAA7362
UAUAAGAGCCACCAUGGACCGUUCACUGGGAUGG
CAAGGGAAUUCUGUCCCUGAGGACAGGACUGAAG
CUGGGAUCAAGCGUUUCCUGGAGGACACCACGGA
UGAUGGAGAACUGAGCAAGUUCGUGAAGGAUUUC
UCAGGAAAUGCGAGCUGCCACCCACCAGAGGCUA
AGACCUGGGCAUCCAGGCCCCAAGUCCCGGAGCCA
AGGCCCCAGGCCCCGGACCUCUAUGAUGAUGACCU
GGAGUUCAGACCCCCCUCGCGGCCCCAGUCCUCUG
ACAACCAGCAGUACUUCUGUGCCCCAGCCCCUCUC
AGCCCAUCUGCCAGGCCCCGCAGCCCAUGGGGCAA
GCUUGAUCCCUAUGAUUCCUCUGAGGAUGACAAG
GAGUAUGUGGGCUUUGCAACCCUCCCCAACCAAG
UCCACCGAAAGUCCGUGAAGAAAGGCUUUGACUU
UACCCUCAUGGUGGCAGGAGAGUCUGGCCUGGGC
AAAUCCACACUUGUCAAUAGCCUCUUCCUCACUG
AUCUGUACCGGGACCGGAAACUUCUUGGUGCUGA
AGAGAGGAUCAUGCAAACUGUGGAGAUCACUAAG
CAUGCAGUGGACAUAGAAGAGAAGGGUGUGAGGC
UGCGGCUCACCAUUGUGGACACACCAGGUUUUGG
GGAUGCAGUCAACAACACAGAGUGCUGGAAGCCU
GUGGCAGAAUACAUUGAUCAGCAGUUUGAGCAGU
AUUUCCGAGACGAGAGUGGCCUGAACCGAAAGAA
CAUCCAAGACAACAGGGUGCACUGCUGCCUGUAC
UUCAUCUCACCCUUCGGCCAUGGGCUCCGGCCAUU
GGAUGUUGAAUUCAUGAAGGCCCUGCAUCAGCGG
GUCAACAUCGUGCCUAUCCUGGCUAAGGCAGACA
CACUGACACCUCCCGAAGUGGACCACAAGAAACGC
AAAAUCCGGGAGGAGAUUGAGCAUUUUGGAAUCA
AGAUCUAUCAAUUCCCAGACUGUGACUCUGAUGA
GGAUGAGGACUUCAAAUUGCAGGACCAAGCCCUA
AAGGAAAGCAUCCCAUUUGCAGUAAUUGGCAGCA
ACACUGUAGUAGAGGCCAGAGGGCGGCGAGUUCG
GGGUCGACUCUACCCCUGGGGCAUCGUGGAAGUG
GAAAACCCAGGGCACUGCGACUUUGUGAAGCUGA
GGACAAUGCUGGUACGUACCCACAUGCAGGACCU
GAAGGAUGUGACACGGGAGACACAUUAUGAGAAC
UACCGGGCACAGUGCAUCCAGAGCAUGACCCGCCU
GGUGGUGAAGGAACGGAAUCGCAACAAACUGACU
CGGGAAAGUGGUACCGACUUCCCCAUCCCUGCUG
UCCCACCAGGGACAGAUCCAGAAACUGAGAAGCU
UAUCCGAGAGAAAGAUGAGGAGCUGCGGCGGAUG
CAGGAGAUGCUACACAAAAUACAAAAACAGAUGA
AGGAGAACUAUUGAUAAUAGGCUGGAGCCUCGGU
GGCCAUGCUUCUUGCCCCUUGGGCCUCCCCCCAGC
CCCUCCUCCCCUUCCUGCACCCGUACCCCCGUGGU
CUUUGAAUAAAGUCUGAGUGGGCGGC
MDRSLGWQGNSVPEDRTEAGIKRFLEDTTDDGELSKF7493
VKDFSGNASCHPPEAKTWASRPQVPEPRPQAPDLYDD
DLEFRPPSRPQSSDNQQYFCAPAPLSPSARPRSPWGKL
DPYDSSEDDKEYVGFATLPNQVHRKSVKKGFDFTLM
VAGESGLGKSTLVNSLFLTDLYRDRKLLGAEERIMQT
VEITKHAVDIEEKGVRLRLTIVDTPGFGDAVNNTECW
KPVAEYIDQQFEQYFRDESGLNRKNIQDNRVHCCLYF
ISPFGHGLRPLDVEFMKALHQRVNIVPILAKADTLTPP
EVDHKKRKIREEIEHFGIKIYQFPDCDSDEDEDFKLQD
QALKESIPFAVIGSNTVVEARGRRVRGRLYPWGIVEV
ENPGHCDFVKLRTMLVRTHMQDLKDVTRETHYENY
RAQCIQSMTRLVVKERNRNKLTRESGTDFPIPAVPPGT
DPETEKLIREKDEELRRMQEMLHKIQKQMKENY
Diablo,GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAA7494
IAP-bindingUAUAAGAGCCACCAUGGCGGCUCUGAAGAGUUGG
mitochondrialCUGUCGCGCAGCGUAACUUCAUUCUUCAGGUACA
proteinGACAGUGUUUGUGUGUUCCUGUUGUGGCUAACUU
(DIABLO)UAAGAAGCGGUGUUUCUCAGAAUUGAUAAGACCA
UGGCACAAAACUGUGACGAUUGGCUUUGGAGUAA
CCCUGUGUGCGGUUCCUAUUGCACAGAAAUCAGA
GCCUCAUUCCCUUAGUAGUGAAGCAUUGAUGAGG
AGAGCAGUGUCUUUGGUAACAGAUAGCACCUCUA
CCUUUCUCUCUCAGACCACAUAUGCGUUGAUUGA
AGCUAUUACUGAAUAUACUAAGGCUGUUUAUACC
UUAACUUCUCUUUACCGACAAUAUACAAGUUUAC
UUGGGAAAAUGAAUUCAGAGGAGGAAGAUGAAGU
GUGGCAGGUGAUCAUAGGAGCCAGAGCUGAGAUG
ACUUCAAAACACCAAGAGUACUUGAAGCUGGAAA
CCACUUGGAUGACUGCAGUUGGUCUUUCAGAGAU
GGCAGCAGAAGCUGCAUAUCAAACUGGCGCAGAU
CAGGCCUCUAUAACCGCCAGGAAUCACAUUCAGC
UGGUGAAACUGCAGGUGGAAGAGGUGCACCAGCU
CUCCCGGAAAGCAGAAACCAAGCUGGCAGAAGCA
CAGAUAGAAGAGCUCCGUCAGAAAACACAGGAGG
AAGGGGAGGAGCGGGCUGAGUCGGAGCAGGAGGC
CUACCUGCGUGAGGAUUGAUAAUAGGCUGGAGCC
UCGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCCC
CCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCC
GUGGUCUUUGAAUAAAGUCUGAGUGGGCGGC
MAALKSWLSRSVTSFFRYRQCLCVPVVANFKKRCFSE1986
LIRPWHKTVTIGFGVTLCAVPIAQKSEPHSLSSEALMR
RAVSLVTDSTSTFLSQTTYALIEAITEYTKAVYTLTSLY
RQYTSLLGKMNSEEEDEVWQVIIGARAEMTSKHQEY
LKLETTWMTAVGLSEMAAEAAYQTGADQASITARNH
IQLVKLQVEEVHQLSRKAETKLAEAQIEELRQKTQEE
GEERAESEQEAYLRED

Peptide fragments identified for the evaluated proteins are shown in Table 43.

TABLE 43
Protein and Peptide Fragment Sequences
Peptide5mC5mCs2U
Fragmentandandand
SEQ ID NOpU1mpU5mCpU1mpU
AIFM1
DGEQHEDLNEV7495YES
AK
TGGLEIDSDFGG7496YESYESYES
FR
COMMD1
ESLMNQSR7497YESYESYESYESYES
HSAQIHTPVAIIE7498YESYESYES
LELGK
WNSGLR7499YESYESYESYES
SEPT4
ESGTDFPIPAVPP7500YESYESYESYESYES
GTDPETEK
FLEDTTDDGELS7501YESYESYESYESYES
K
HAVDIEEK7502YESYESYESYESYES
DIABLO
AVYTLTSLYR7503YESYESYESYESYES
LAEAQIEELR7504YESYESYESYESYES
NHIQLVK7505YESYESYESYESYES

Human lung cancer A549 cells were plated in 6-wells, and transfected with Lipofectamine 2000 (Life Technologies) and 5 μg of constitutively active (C.A.) caspase 3 mRNA (mRNA sequence shown in SEQ ID NO: 6619 (Table 44); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1) or constitutively active (C.A.) caspase 6 mRNA (mRNA sequence shown in SEQ ID NO: 7506 (Table 44); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1 mpU) or fully modified with 1-methylpseudouridine (1mpU). Cells were harvested 7-10 hours post-transfection and lysed in RIPA buffer containing a protease inhibitor co*cktail (Roche, Indianapolis, Ind.). 20 μg of cell lysate per lane was run for Western blotting to detect endogenous and introduced caspase 3; endogenous and introduced caspase 6; the caspase 3 downstream-substrate PARP; and the caspase 6 downstream-substrate lamin A/C. Compared to control lysate, higher levels of cleaved caspase 3 and cleaved caspase 6 were detected in C.A. caspase 3 and C.A. caspase 6 modified mRNA transfected cells, respectively. As shown in FIG. 7, cleavage of the downstream substrates PARP and lamin A/C were detected in cells treated with C.A. caspase 3 modified mRNA (FIG. 7A) and C.A. caspase 6 modified mRNAs (FIG. 7B). The 5 lanes of the Westerns shown in FIGS. 7A and 7B contain lysate from the following: 1) untransfected HeLa cells, 2) untransfected A549, 3) A549 lipofectamine alone control, 4) A549 transfected with 5mC, 1mpU modified mRNA, and 5) A549 transfected with 1 mpU modified mRNA.

TABLE 44
C.A. Caspase Sequences
SEQ ID
DescriptionSequenceNO:
C.A.GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUA6619
caspase 3UAAGAGCCACCAUGAUUGAGACAGACAGUGGUGUUGA
UGAUGACAUGGCGUGUCAUAAAAUACCAGUGGAGGCC
GACUUCUUGUAUGCAUACUCCACAGCACCUGGUUAUU
AUUCUUGGCGAAAUUCAAAGGAUGGCUCCUGGUUCAU
CCAGUCGCUUUGUGCCAUGCUGAAACAGUAUGCCGAC
AAGCUUGAAUUUAUGCACAUUCUUACCCGGGUUAACC
GAAAGGUGGCAACAGAAUUUGAGUCCUUUUCCUUUGA
CGCUACUUUUCAUGCAAAGAAACAGAUUCCAUGUAUU
GUUUCCAUGCUCACAAAAGAACUCUAUUUUUAUCACG
AUGAAGUUGAUGGGGGAUCCCCCAUGGAGAACACUGA
AAACUCAGUGGAUUCAAAAUCCAUUAAAAAUUUGGA
ACCAAAGAUCAUACAUGGAAGCGAAUCAAUGGACUCU
GGAAUAUCCCUGGACAACAGUUAUAAAAUGGAUUAUC
CUGAGAUGGGUUUAUGUAUAAUAAUUAAUAAUAAGA
AUUUUCAUAAGAGCACUGGAAUGACAUCUCGGUCUGG
UACAGAUGUCGAUGCAGCAAACCUCAGGGAAACAUUC
AGAAACUUGAAAUAUGAAGUCAGGAAUAAAAAUGAU
CUUACACGUGAAGAAAUUGUGGAAUUGAUGCGUGAU
GUUUCUAAAGAAGAUCACAGCAAAAGGAGCAGUUUU
GUUUGUGUGCUUCUGAGCCAUGGUGAAGAAGGAAUA
AUUUUUGGAACAAAUGGACCUGUUGACCUGAAAAAA
AUAACAAACUUUUUCAGAGGGGAUCGUUGUAGAAGU
CUAACUGGAAAACCCAAACUUUUCAUUAUUCAGGCCU
GCCGUGGUACAGAACUGGACUGUGGCAUUGAGACAGA
CUGAUAAUAGGCUGGAGCCUCGGUGGCCAUGCUUCUU
GCCCCUUGGGCCUCCCCCCAGCCCCUCCUCCCCUUCCU
GCACCCGUACCCCCGUGGUCUUUGAAUAAAGUCUGAG
UGGGCGGC
MIETDSGVDDDMACHKIPVEADFLYAYSTAPGYYSW2484
RNSKDGSWFIQSLCAMLKQYADKLEFMHILTRVNRK
VATEFESFSFDATFHAKKQIPCIVSMLTKELYFYHDE
VDGGSPMENTENSVDSKSIKNLEPKIIHGSESMDSGIS
LDNSYKMDYPEMGLCIIINNKNFHKSTGMTSRSGTD
VDAANLRETFRNLKYEVRNKNDLTREEIVELMRDVS
KEDHSKRSSFVCVLLSHGEEGIIFGTNGPVDLKKITNF
FRGDRCRSLTGKPKLFIIQACRGTELDCGIETD
C.A.GGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAA7506
caspase 6AUAUAAGAGCCACCAUGGUAGAAAUAGAUGCAGC
CUCCGUUUACACGCUGCCUGCUGGAGCUGACUUC
CUCAUGUGUUACUCUGUUGCAGAAGGAUAUUAU
UCUCACCGGGAAACUGUGAACGGCUCAUGGUACA
UUCAAGAUUUGUGUGAGAUGUUGGGAAAAUAUG
GCUCCUCCUUAGAGUUCACAGAACUCCUCACACU
GGUGAACAGGAAAGUUUCUCAGCGCCGAGUGGAC
UUUUGCAAAGACCCAAGUGCAAUUGGAAAGAAGC
AGGUUCCCUGUUUUGCCUCAAUGCUAACUAAAAA
GCUGCAUUUCUUUCCAAAAUCUAAUCUCGAGCAC
CACCACCACCACCACGUUGAAAUUGAUGGGGGAU
CCCCCAUGAGCUCGGCCUCGGGGCUCCGCAGGGG
GCACCCGGCAGGUGGGGAAGAAAACAUGACAGAA
ACAGAUGCCUUCUAUAAAAGAGAAAUGUUUGAU
CCGGCAGAAAAGUACAAAAUGGACCACAGGAGGA
GAGGAAUUGCUUUAAUCUUCAAUCAUGAGAGGU
UCUUUUGGCACUUAACACUGCCAGAAAGGCGGGG
CACCUGCGCAGAUAGAGACAAUCUUACCCGCAGG
UUUUCAGAUCUAGGAUUUGAAGUGAAAUGCUUU
AAUGAUCUUAAAGCAGAAGAACUACUGCUCAAAA
UUCAUGAGGUGUCAACUGUUAGCCACGCAGAUGC
CGAUUGCUUUGUGUGUGUCUUCCUGAGCCAUGGC
GAAGGCAAUCACAUUUAUGCAUAUGAUGCUAAA
AUCGAAAUUCAGACAUUAACUGGCUUGUUCAAAG
GAGACAAGUGUCACAGCCUGGUUGGAAAACCCAA
GAUAUUUAUCAUCCAGGCAUGUCGGGGAAACCAG
CACGAUGUGCCAGUCAUUCCUUUGGAUGUAGUAG
AUUGAUAAUAGGCUGGAGCCUCGGUGGCCAUGCU
UCUUGCCCCUUGGGCCUCCCCCCAGCCCCUCCUCC
CCUUCCUGCACCCGUACCCCCGUGGUCUUUGAAU
AAAGUCUGAGUGGGCGGC
MVEIDAASVYTLPAGADFLMCYSVAEGYYSHRETVN2486
GSWYIQDLCEMLGKYGSSLEFTELLTLVNRKVSQRR
VDFCKDPSAIGKKQVPCFASMLTKKLHFFPKSNLEHH
HHHHVEIDGGSPMSSASGLRRGHPAGGEENMTETDA
FYKREMFDPAEKYKMDHRRRGIALIFNHERFFWHLT
LPERRGTCADRDNLTRRFSDLGFEVKCFNDLKAEELL
LKIHEVSTVSHADADCFVCVFLSHGEGNHIYAYDAKI
EIQTLTGLFKGDKCHSLVGKPKIFIIQACRGNQHDVPV
IPLDVVD

The activity of cultured human lung adenocarcinoma A549 cells was evaluated through the measurement of formazan converted by mitochondrial dehydrogenases from WST-1 substrate (Roche, Indianapolis, Ind.). 7500 cells per 96-well were treated with a single dose of varying amounts of Lipofectamine 2000-lipoplexed constitutively active (C.A.) caspase 3 mRNA (mRNA sequence shown in SEQ ID NO: 6619 (Table 44); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) or constitutively active (C.A.) caspase 6 mRNA (mRNA sequence shown in SEQ ID NO: 7506 (Table 44); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1 mpU) or fully modified with 1-methylpseudouridine (1 mpU) or a control proteins (eGFP (mRNA sequence shown in SEQ ID NO: 7507; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1 mpU) or fully modified with 1-methylpseudouridine (1mpU)) and luciferase (mRNA sequence shown in SEQ ID NO: 7508; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1 mpU) or fully modified with 1-methylpseudouridine (1mpU))). Cellular activity was measured in cultured cells 1 day after mRNA treatment according to the WST-1 manufacturer's protocol, and is plotted as a 450 nm absorbance reading of the converted formazan that had been corrected for background signal. As shown in Table 45, increasing amounts of transfected C.A. caspase mRNA (0 ng, 2 ng, 10 ng, 50 ng and 250 ng) markedly inhibited the optical density (OD) signal (as a readout of cellular activity) compared to controls. Similar results were obtained in human lung adenocarcinoma H441 cells and human cervical cancer HeLa cells.

TABLE 45
Cellular Activity
Amount of mRNAWST-1 OD mean
Description(ng)(450-690 nm)
C.A. caspase 302.46
(1mpU and 5mC)21.93
101.05
500.31
2500.04
C.A. caspase 602.49
(1mpU and 5mC)22.41
101.64
500.75
2500.30
eGFP02.37
(1mpU and 5mC)22.36
102.19
501.84
2501.84
Luciferase02.26
(1mpU and 5mC)21.93
102.00
501.94
2501.87
C.A. caspase 302.62
(1mpU)22.35
101.80
500.91
2500.17
C.A. caspase 602.17
(1mpU)22.34
102.01
501.29
2500.42
eGFP02.56
(1mpU)22.67
102.86
502.72
2502.38
Luciferase02.12
(1mpU)22.56
102.68
502.64
2502.21

Human hepatocellular carcinoma Hep3B cells were plated in a 6-well plate at a seeding density of 3×106 cells/well and Lipofectamine 2000-transfected with mRNAs fully modified with 5-methylcytidine and 1-methylpseudouridine (5mC and 1 mpU) or fully modified with 1-methylpseudouridine (1mpU) designed to encode the following: fluorescent protein mCherry (mRNA sequence shown in SEQ ID NO: 6602; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), non-translatable Factor 1× (mRNA sequence shown in SEQ ID NO: 7509; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1), full length wildtype C-MYC (mRNA sequence shown in SEQ ID NO: 7510; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), MYC inhibitor A (mRNA sequence shown in SEQ ID NO: 7511 (Table 46); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), MYC inhibitor B (mRNA sequence shown in SEQ ID NO: 7513 (Table 46); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1), MYC inhibitor C (mRNA sequence shown in SEQ ID NO: 7418 (Table 46); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) and MYC inhibitor D (mRNA sequence shown in SEQ ID NO: 7515 (Table 46); polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1). Cells were collected 8 hours post-transfection and lysates were made using RIPA lysis buffer including a protease inhibitor co*cktail (Roche, Indianapolis, Ind.). Equal amounts of lysate determined by BCA assay were resolved by SDS-PAGE through 4-12% BIS-TRIS gels, transferred to nitrocellulose blots and probed with appropriate primary and secondary antibodies. Western blot analyses revealed positive expression of the 4 modified mRNA MYC inhibitors, as well as full length C-MYC, in Hep3B cells.

TABLE 46
MYC Inhibitor Sequences
SEQ ID
DescriptionSequenceNO:
MYCGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAA7511
inhibitor AAUAUAAGAGCCACCAUGACCGAAGAAAACGUCAA
GAGAAGAACCCAUAAUGUCCUCGAGCGCCAGCGG
CGCAAUGAGCUCAAGCGCAGCUUCUUUGCACUCA
GGGACCAAAUUCCAGAGUUGGAGAACAACGAAAA
GGCCCCGAAGGUGGUGAUCCUUAAGAAGGCGACU
GCCUACAUCCUGUCGGUGCAGGCUGAGACUCAAA
AGCUGAUCUCCGAAAUCGAUCUGCUCCGGAAACA
GAACGAACAACUGAAACACAAACUGGAACAGCUG
CGGAAUUCAUGCGCGUGAUAAUAGGCUGGAGCCU
CGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCCC
CCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCC
GUGGUCUUUGAAUAAAGUCUGAGUGGGCGGC
MTEENVKRRTHNVLERQRRNELKRSFFALRDQIPELE7512
NNEKAPKVVILKKATAYILSVQAETQKLISEIDLLRKQ
NEQLKHKLEQLRNSCA
MYCGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAA7513
inhibitor BAUAUAAGAGCCACCAUGACCGAAGAAAACGUCAA
GAGAAGAACCCAUAAUGUCCUCGAGCGCCAGCGG
CGCAAUGAGCUCAAGCGCAGCUUCUUUGCACUCA
GGGACCAAAUUCCAGAGUUGGAGAACAACGAAAA
GGCCCCGAAGGUGGUGAUCCUUAAGAAGGCGACU
GCCUACAUCCUGUCGGUGCAGGCUGAGAAUCAAA
AGCUGAUCUCCGAAAUCGAUCUGCUCCGGAAACA
GAACGAACAACUGAAACACAAACUGGAACAGCUG
CGGAAUUCAUGCGCGUGAUAAUAGGCUGGAGCCU
CGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCCC
CCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCC
GUGGUCUUUGAAUAAAGUCUGAGUGGGCGGC
MTEENVKRRTHNVLERQRRNELKRSFFALRDQIPELE7514
NNEKAPKVVILKKATAYILSVQAENQKLISEIDLLRK
QNEQLKHKLEQLRNSCA
MYCGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAA7515
inhibitor CAUAUAAGAGCCACCAUGAGCGCCGCUGAUAAGCG
GGCUCACCACAAUGCGUUGGAGAGGAAGAGGCGC
GACCACAUCAAAGACUCGUUCCAUUCACUCCGGG
ACUCCGUGCCGUCGCUGCAAGGAGAAAAAGCCUC
CCGGGCACAGAUCCUCGACAAGGCGACUGAGUAC
AUUCAGUACAUGCGCCGCAAGAACCACACCCAUC
AGCAAGAUAUCGACGAUCUUAAGAGACAGAACGC
GCUGCUGGAACAACAGGUCCGCGCACUGGAAAAG
GCCAGAAGCUCAGCCUGAUAAUAGGCUGGAGCCU
CGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCCC
CCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCC
GUGGUCUUUGAAUAAAGUCUGAGUGGGCGGC
MSAADKRAHHNALERKRRDHIKDSFHSLRDSVPSLQ7516
GEKASRAQILDKATEYIQYMRRKNHTHQQDIDDLKR
QNALLEQQVRALEKARSSA
MYCGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUA6621
inhibitor DUAAGAGCCACCAUGACCGAAGAAAACGUCAAGAGAAG
AACCCAUAAUGUCCUCGAGCGCCAGCGGCGCAAUGAG
CUCAAGCGCAGCUUCUUUGCACUCAGGGACCAAAUUC
CAGAGUUGGAGAACAACGAAAAGGCCCCGAAGGUGGU
GAUCCUUAAGAAGGCGACUGCCUACAUCCUGUCGGUG
CAGGCUGAGACUCAAAAGCUGAUCUCCGAAAUCGAUC
UGCUCCGGAAACAGAACGAACAACUGAAACACAAACU
GGAACAGCUGCGGAAUUCAUGCUGAUAAUAGGCUGGA
GCCUCGGUGGCCAUGCUUCUUGCCCCUUGGGCCUCCC
CCCAGCCCCUCCUCCCCUUCCUGCACCCGUACCCCCGU
GGUCUUUGAAUAAAGUCUGAGUGGGCGGC
MTEENVKRRTHNVLERQRRNELKRSFFALRDQIPELE7517
NNEKAPKVVILKKATAYILSVQAETQKLISEIDLLRKQ
NEQLKHKLEQLRNSC

Hep3B cells were plated in a 96-well plate at a seeding density of 2500 cells/well and Lipofectamine 2000-transfected with 0, 0.2 nM, 0.7 nM, 2 nM or 6 nM of modified mRNAs fully modified with 1-methylpseudouridine (1mpU) designed to encode the following: mCherry (mRNA sequence shown in SEQ ID NO: 6602; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), non-translatable Factor 1× (mRNA sequence shown in SEQ ID NO: 7509; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), full length wildtype C-MYC (mRNA sequence shown in SEQ ID NO: 7510; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), MYC inhibitor A (mRNA sequence shown in SEQ ID NO: 7511; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), MYC inhibitor B (mRNA sequence shown in SEQ ID NO: 7513; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1), MYC inhibitor C (mRNA sequence shown in SEQ ID NO: 7515; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1) and MYC inhibitor D (mRNA sequence shown in SEQ ID NO: 6621; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1). Cellular activity was measured 48 hours post-transfection with the use of WST-1 according to manufacturer's instructions (Roche, Indianapolis, Ind.). Absorbance readings were taken at 450 nm 4 hours after the addition of WST-1, and background-corrected results are shown in Table 47. The three highest concentrations of each of the inhibitors (MYC inhibitor A, MYC inhibitor B, MYC inhibitor C and MYC inhibitor D) reduced absorbance signal compared to the controls.

TABLE 47
Cellular Activity
Amount of mRNAWST-1 OD mean
Description(nM)(450-690 nm)
MYC inhibitor A00.45
0.20.42
0.70.09
20.12
60.05
MYC inhibitor B00.67
0.20.69
0.70.24
20.09
60.05
MYC inhibitor C00.73
0.20.73
0.70.34
20.09
60.04
MYC inhibitor D00.74
0.20.68
0.70.32
20.14
60.07
mCherry00.66
0.20.66
0.70.62
20.60
60.51
Non-translatable00.65
FIX0.20.65
0.70.61
20.62
60.49
Wild-Type MYC00.58
0.20.51
0.70.51
20.51
60.46

A. BALB/C Nude Mice

BALB/c nude mice were injected intravenously with 0.1 mg/kg luciferase modified mRNA without a miR-122 binding site (“non-targeted mRNA”; mRNA sequence shown in SEQ ID NO: 7518; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and 1-methylpseudouridine) formulated in a lipid nanoparticle described in Table 48 or luciferase modified mRNA with a miR-122 binding site in the 3′UTR (“miR-122 targeted mRNA”; mRNA sequence shown in SEQ ID NO: 7519; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1; fully modified with 5-methylcytidine and 1-methylpseudouridine) formulated in a lipid nanoparticle described in Table 49.

TABLE 48
Lipid Nanoparticle for Non-targeted mRNA
LNPLuciferase: non-targeted mRNA
LipidDLin-KC2-DMA
Lipid/RNA wt/wt20
Mean size73.3 nm
PDI: 0.06
TABLE 49
Lipid Nanoparticle for Targeted mRNA
LNPLuciferase: targeted mRNA
LipidDLin-KC2-DMA
Lipid/RNA wt/wt20
Mean size70.6 nm
PDI: 0.08

24 hours post-treatment, animals were anesthetized, injected with the luciferase substrate D-luciferin and the bioluminescence imaging (BLI) from living animals was evaluated in an IVIS imager 15 minutes later. Signals were obtained from animals injected with non-targeted mRNA and from miR-122 targeted mRNA, and presented in Table 50. The total light signal produced from livers of animals treated with miR 122 targeted mRNA is 29× lower than non-targeted mRNA, showing that the engineered element in the 3′UTR may inhibit protein expression in normal tissue.

TABLE 50
In vivo expression of modified mRNA modulated by
an engineered miR122 binding site
Luciferase signal from liver
Description(photons/sec)
Non-targeted mRNA7.9 × 107
miR-122 targted mRNA2.7 × 106

B. BALB/c Nude Mice with Hepatocellular Carcinoma Hep3B Cells

BALB/c nude mice were intrahepatic ally implanted with 2×106 hepatocellular carcinoma Hep3B cells and resulting orthotopic tumors allowed to grow for 24 days. Tumor-bearing mice were then intravenously injected with 0.1 mg/kg luciferase modified mRNA without a miR-122 binding site (“non-targeted mRNA”; mRNA sequence shown in SEQ ID NO: 7518; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and 1-methylpseudouridine) or luciferase modified mRNA with a miR-122 binding site in the 3′UTR (“miR-122 targeted mRNA”; mRNA sequence shown in SEQ ID NO: 7519; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytidine and 1-methylpseudouridine) formulated in a lipid nanoparticle described in Table 45 (above). 24 hr post-treatment animals were anesthetized, injected with the luciferase substrate D-luciferin and bioluminescence imaging (BLI) from living animals was evaluated in an IVIS imager 20 minutes later. Signal from orthotopic tumors compared to adjacent normal liver was quantified, and miR-122-targeted mRNA systemically delivered via lipid nanoparticles achieved over 2-fold enrichment in tumor compared to normal liver.

A. HeLa Cells

HeLa cells were seeded at a density of 15,000 per well in 100 ul cell culture medium (DMEM+10% FBS). G-CSF mRNA having a miR-122 sequence in the 3′UTR (G-CSF miR122; mRNA sequence shown in SEQ ID NO: 7325; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap 1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or G-CSF mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF seedless; mRNA sequence shown in SEQ ID NO: 7327; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 0.3 ul per well of Lipofectamine 2000 at a concentration of 75 ng of mRNA per well in 96 well plates. The supernatant was collected between 16-18 hours after transfection and expression of G-CSF was measured by ELISA, and the results are shown in Table 51.

TABLE 51
G-CSF Expression in HeLa
Protein Expression
Description(ng/ml)
G-CSF miR122292.1
G-CSF seedless335.7

B. Primary Human and Rat Hepatocytes

Primary human or rat hepatocytes cells were seeded at a density of 350,000 cells per well in 500 ul cell culture medium (InvitroGRO CP and InVitroGRO HI Medium+2.2% Torpedo Antibiotic Mix). G-CSF mRNA having a miR-122 sequence in the 3′UTR (G-CSF miR122; mRNA sequence shown in SEQ ID NO: 7520; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or G-CSF mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF seedless; mRNA sequence shown in SEQ ID NO: 7521; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 1 ul per well of Lipofectamine 2000 at a concentration of 500 ng of mRNA per well in 24 well plates for the primary human hepatocytes and the primary rat hepatocytes. The supernatant was collected between 16-18 hours after transfection and expression of G-CSF was measured by ELISA, and the results are shown in Table 52. The mir-122 binding site sequence in the mRNA dampened the G-CSF protein expression in the primary hepatocytes.

TABLE 52
G-CSF Expression in Hepatocytes
Primary Human
HepatocytesPrimary Rat Hepatocytes
DescriptionProtein Expression (ng/ml)Protein Expression (ng/ml)
G-CSF miR12211626
G-CSF seedless46385

A. HeLa Cells

HeLa cells were seeded at a density of 17,000 per well in 100 ul cell culture medium (DMEM+10% FBS). G-CSF mRNA without a miR-122 sequence in the 3′UTR (G-CSF; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7321; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7320; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 sequence in the 3′UTR (G-CSF miR122; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7325; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7322; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 seed sequence in the 3′UTR (G-CSF seed; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7326; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7323; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF seedless; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7327; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7324; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), Factor 1× mRNA without a miR-122 sequence in the 3′UTR (FIX; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7329; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7328; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), Factor 1× mRNA having a miR-122 sequence in the 3′UTR (FIX miR122; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7333; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7330; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), Factor 1× mRNA having a miR-122 seed sequence in the 3′UTR (FIX seed; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7334; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7331; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or Factor 1× mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (FIX seedless; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7335; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7332; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 0.3 ul per well of Lipofectamine 2000 at a concentration of 75 ng of mRNA per well in 96 well plates. The supernatant was collected between 16-18 hours after transfection, expression of G-CSF or Factor 1× was measured by ELISA, and the results are shown in Table 53.

TABLE 53
Expression in HeLa
Protein ExpressionProtein Expression
Mm 3′UTRHs 3′UTR
Description(ng/ml)(ng/ml)
G-CSF271.7269.4
G-CSF miR122305.3668.8
G-CSF seed209.598.0
G-CSF seedless243.280.9
FIX249.8131.6
FIX mir122204.655.4
FIX seed290.05127.6
FIX seedless180.931.6

B. Primary Human and Rat Hepatocytes

Primary human or rat hepatocytes cells were seeded at a density of 350,000 cells per well in 500 ul cell culture medium (InvitroGRO CP and InVitroGRO HI Medium+2.2% Torpedo Antibiotic). G-CSF mRNA without a miR-122 sequence in the 3′UTR (G-CSF; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7321; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7320; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 sequence in the 3′UTR (G-CSF miR122; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7325; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7322; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 seed sequence in the 3′UTR (G-CSF seed; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7326; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7323; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF seedless; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7327; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7324; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), Factor 1× mRNA without a miR-122 sequence in the 3′UTR (FIX; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7329; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7328; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), Factor 1× mRNA having a miR-122 sequence in the 3′UTR (FIX miR122; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7333; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7330; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), Factor 1× mRNA having a miR-122 seed sequence in the 3′UTR (FIX seed; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7334; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7331; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or Factor 1× mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (FIX seedless; mouse 3′ UTR mRNA sequence shown in SEQ ID NO: 7335; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine; human 3′UTR mRNA sequence shown in SEQ ID NO: 7332; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 1 ul per well of Lipofectamine 2000 at a concentration of 500 ng per well in 24 well plates for the primary human hepatocytes and the primary rat hepatocytes. The supernatant was collected at 24 hours, 48 hours and 72 hours after transfection, expression of G-CSF and Factor 1× was measured by ELISA, and the results are shown in Table 54. The mir-122 binding site sequence in the mRNA dampened the G-CSF and Factor 1× protein expression in the primary hepatocytes.

TABLE 54
G-CSF Expression in Hepatocytes
Primary HumanPrimary Human
HepatocytesHepatocytes
Protein ExpressionProtein Expression
(ng/ml)(ng/ml)
DescriptionTime PointMm 3′UTRHs 3′UTR
G-CSF24 hours43.984.9
48 hours18.8100.4
72 hours5.721.3
G-CSF miR12224 hours6.924.0
48 hours.73.03
72 hours.12.88
G-CSF seed24 hours48.5115.8
48 hours25.696.4
72 hours8.219.2
G-CSF seedless24 hours31.7113.1
48 hours11.792.9
72 hours3.418.9
FIX24 hours90.863.2
48 hours159.6124.8
72 hours70.544.3
FIX mir12224 hours11.815.9
48 hours5.04.4
72 hours1.0.4
FIX seed24 hours77.260.2
48 hours115.063.0
72 hours41.720.1
FIX seedless24 hours69.353.7
48 hours123.875.0
72 hours49.024.5

A. Base Level of miR-122

The base level of mir-122 in Human hepatocytes, rat hepatocytes, human hepatocellular carcinoma cells (Hep3B) and HeLa cells were determined by TAQMAN® analysis using the manufacturers protocol. The levels were normalized to U6 and the results are shown in Table 55.

TABLE 55
miR-122 Levels in Various Cell Types
Cell TypemiR-122 level (normalized to U6)
Human Hepatocytes16.8
Rat Hepatocytes10.9
Hep3B0
HeLa0

B. Primary Human Hepatocytes and Hep3B Cells

Primary human hepatocytes were seeded at a density of 50,000 cells per well in 100 ul cell culture medium (InvitroGRO CP and InVitroGRO HI Medium+2.2% Torpedo Antibiotic Mix) and Hep3B cells were seeded at a density of 20,000 cells per well in 100 ul cell culture medium MEM+10% FBS. G-CSF mRNA without a miR-122 sequence in the 3′UTR (G-CSF; mRNA sequence shown in SEQ ID NO: 7320; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 sequence in the 3′UTR (G-CSF miR122; mRNA sequence shown in SEQ ID NO: 7322; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-122 seed sequence in the 3′UTR (G-CSF seed; mRNA sequence shown in SEQ ID NO: 7323; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or G-CSF mRNA having a miR-122 sequence without the seed sequence in the 3′UTR (G-CSF seedless; mRNA sequence shown in SEQ ID NO: 7324; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 0.3 ul per well of Lipofectamine 2000 at a concentration of 75 ng of mRNA per well in 96 well plates for the primary human hepatocytes and the Hep3B cells. The supernatant was collected at 24 hours, 48 hours and 72 hours after transfection, expression of G-CSF was measured by ELISA, and the results are shown in Table 56. The mir-122 binding site sequence in the mRNA dampened the G-CSF protein expression in the primary human hepatocytes but not in the Hep3B cells.

TABLE 56
G-CSF Expression
Primary Human
HepatocytesHep3B
Protein ExpressionProtein Expression
(ng/ml)(ng/ml)
DescriptionTime PointHs 3′UTRHs 3′UTR
G-CSF24 hours7655
48 hours1233
72 hours610
G-CSF miR12224 hours3237
48 hours127
72 hours06
G-CSF seed24 hours7539
48 hours1128
72 hours46
G-CSF seedless24 hours7949
48 hours1535
72 hours69

A. Base Level of miR-143 0.3p

The base level of miR-142 3p in RAW264.7 cells and HeLa cells were determined by TAQMAN® analysis using the manufacturer's protocol. The levels were normalized to U6 and the results are shown in Table 57.

TABLE 57
miR-142 3p Levels in Various Cell Types
Cell TypemiR-122 level (normalized to U6)
Human Hepatocytes16.8
Rat Hepatocytes10.9
Hep3B0
HeLa0

B. HeLa and RAW264.7 Cells

HeLa cells were seeded at a density of 17,000 per well in 100 ul cell culture medium DMEM+10% FBS and RAW264.7 cells were seeded at a density of 200,000 per well in 100 ul cell culture medium DMEM+10% FBS. G-CSF mRNA without a miR-142 3p sequence in the 3′UTR (G-CSF; mRNA sequence shown in SEQ ID NO: 7522; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-142 3p sequence in the 3′UTR (G-CSF miR142 3p; mRNA sequence shown in SEQ ID NO: 7523; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-142 3p seed sequence in the 3′UTR (G-CSF seed; mRNA sequence shown in SEQ ID NO: 7524; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or G-CSF mRNA having a miR-142 3p sequence without the seed sequence in the 3′UTR (G-CSF seedless; mRNA sequence shown in SEQ ID NO: 7525; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 0.3 ul per well of Lipofectamine 2000 at a concentration of 75 ng of mRNA per well in 96 well plates for HeLa or with 1 ul per well of Lipofectamine 2000 at a concentration of 250 ng of mRNA per well in 24 well plates for RAW264.7 cells. The supernatant was collected 16-18 hours after transfection, expression of G-CSF was measured by ELISA, and the results are shown in Table 58. miR-142 3p sites in G-CSF were shown to down-regulate G-CSF expression in RAW264.7 cells.

TABLE 58
Expression
HeLaRAW264.7
Protein ExpressionProtein Expression
Description(ng/ml)(ng/ml)
G-CSF243.5124.8
G-CSF miR142 3p309.142.8
G-CSF seed259.8148.1
G-CSF seedless321.7185.2

C. Time Course in RAW264.7 Cells

RAW264.7 cells were seeded at a density of 60,000 cells per well in 100 ul cell culture medium (DMEM+10% FBS). G-CSF mRNA without a miR-142 3p sequence in the 3′UTR (G-CSF; mRNA sequence shown in SEQ ID NO: 7522; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-142 3p sequence in the 3′UTR (G-CSF miR142 3p; mRNA sequence shown in SEQ ID NO: 7523; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-142 3p seed sequence in the 3′UTR (G-CSF seed; mRNA sequence shown in SEQ ID NO: 7524; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or G-CSF mRNA having a miR-142 3p sequence without the seed sequence in the 3′UTR (G-CSF seedless; mRNA sequence shown in SEQ ID NO: 7525; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected with 0.3 ul per well of Lipofectamine 2000 at a concentration of 75 ng of mRNA per well in 96 well plates. The supernatant was collected at 24 hours, 48 hours and 72 hours after transfection, expression of G-CSF was measured by ELISA, and the results are shown in Table 59. The mir-142 3p binding site sequence in the mRNA showed a strong suppression of G-CSF expression in RAW264.7 cells over time.

TABLE 59
G-CSF Expression
RAW264.7 Cells
DescriptionTime PointProtein Expression (ng/ml)
G-CSF24 hours133.5
48 hours69.7
72 hours2.1
G-CSF miR142 3p24 hours60.1
48 hours9.2
72 hours.3
G-CSF seed24 hours244.9
48 hours68.9
72 hours2.3
G-CSF seedless24 hours250.2
48 hours95.9
72 hours3.0

D. miR-142 3p in PBMC

Peripheral blood mononuclear cells (PBMCs) were seeded at a density of 150,000 cells per well in 100 ul cell culture medium (Opti-MEM and after transfection add 10% FBS). G-CSF mRNA having a miR-142 3p sequence in the 3′UTR (G-CSF miR142 3p; mRNA sequence shown in SEQ ID NO: 7523; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, Cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine), G-CSF mRNA having a miR-142 3p seed sequence in the 3′UTR (G-CSF seed; mRNA sequence shown in SEQ ID NO: 7524; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) or G-CSF mRNA having a miR-142 3p sequence without the seed sequence in the 3′UTR (G-CSF seedless; mRNA sequence shown in SEQ ID NO: 7525; polyA tail of approximately 140 nucleotides not shown in sequence; 5′ cap, cap1; fully modified with 5-methylcytosine and 1-methylpseudouridine) were transfected in triplicate with 0.4 ul per well of Lipofectamine 2000 at a concentration of 500 ng of mRNA per well in 96 well plates for 2 or 3 donors. The supernatant was collected at 24 hours after transfection and the expression of G-CSF was measured by ELISA. The results for the 2 donors are shown in Table 60 and the results for the 3 donors are shown in Table 61. The mir-142 3p binding site sequence in the mRNA was shown to down regulate G-CSF expression in human PBMC.

TABLE 60
Expression PBMC (2 donors)
DescriptionProtein Expression (ng/ml)
G-CSF miR142 3p5.09
G-CSF seed10.06
G-CSF seedless9.38
TABLE 61
Expression PBMC (3 donors)
DescriptionProtein Expression (ng/ml)
G-CSF miR142 3p7.48
G-CSF seed13.40
G-CSF seedless13.98

It is to be understood that the words which have been used are words of description rather than limitation, and that changes may be made within the purview of the appended claims without departing from the true scope and spirit of the invention in its broader aspects.

While the present invention has been described at some length and with some particularity with respect to the several described embodiments, it is not intended that it should be limited to any such particulars or embodiments or any particular embodiment, but it is to be construed with references to the appended claims so as to provide the broadest possible interpretation of such claims in view of the prior art and, therefore, to effectively encompass the intended scope of the invention.

All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, section headings, the materials, methods, and examples are illustrative only and not intended to be limiting.

Signal-sensor polynucleotides for the alteration of cellular phenotypes (2024)
Top Articles
Latest Posts
Article information

Author: Aron Pacocha

Last Updated:

Views: 6055

Rating: 4.8 / 5 (48 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Aron Pacocha

Birthday: 1999-08-12

Address: 3808 Moen Corner, Gorczanyport, FL 67364-2074

Phone: +393457723392

Job: Retail Consultant

Hobby: Jewelry making, Cooking, Gaming, Reading, Juggling, Cabaret, Origami

Introduction: My name is Aron Pacocha, I am a happy, tasty, innocent, proud, talented, courageous, magnificent person who loves writing and wants to share my knowledge and understanding with you.