US20200261599A1 - Nucleic acid products and methods of administration thereof - Google Patents

Nucleic acid products and methods of administration thereof Download PDF

Info

Publication number
US20200261599A1
US20200261599A1 US16/789,831 US202016789831A US2020261599A1 US 20200261599 A1 US20200261599 A1 US 20200261599A1 US 202016789831 A US202016789831 A US 202016789831A US 2020261599 A1 US2020261599 A1 US 2020261599A1
Authority
US
United States
Prior art keywords
cancer
cell
protein
seq
human
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US16/789,831
Inventor
Matthew Angel
Christopher Rohde
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Factor Bioscience Inc
Original Assignee
Factor Bioscience Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Factor Bioscience Inc filed Critical Factor Bioscience Inc
Priority to US16/789,831 priority Critical patent/US20200261599A1/en
Assigned to FACTOR BIOSCIENCE INC. reassignment FACTOR BIOSCIENCE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANGEL, MATTHEW, ROHDE, CHRISTOPHER
Publication of US20200261599A1 publication Critical patent/US20200261599A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/02Peptides of undefined number of amino acids; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0015Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin by using microneedles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/20Chemical, physico-chemical or functional or structural properties of the composition as a whole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M2037/0007Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin having means for enhancing the permeation of substances through the epidermis, e.g. using suction or depression, electric or magnetic fields, sound waves or chemical agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0015Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin by using microneedles
    • A61M2037/0023Drug applicators using microneedles

Definitions

  • the present invention relates, in part, to methods, compositions, and products for producing and delivering nucleic acids to cells, tissues, organs, and patients, methods for expressing proteins in cells, tissues, organs, and patients, and cells, therapeutics, and cosmetics produced using these methods, compositions, and products.
  • RNA Ribonucleic acid
  • Ribonucleic acid is ubiquitous in both prokaryotic and eukaryotic cells, where it encodes genetic information in the form of messenger RNA, binds and transports amino acids in the form of transfer RNA, assembles amino acids into proteins in the form of ribosomal RNA, and performs numerous other functions including gene expression regulation in the forms of microRNA and long non-coding RNA.
  • RNA can be produced synthetically by methods including direct chemical synthesis and in vitro transcription, and can be administered to patients for therapeutic use.
  • previously described synthetic RNA molecules are unstable and trigger a potent innate-immune response in human cells.
  • methods for efficient non-viral delivery of nucleic acids to patients, organs, tissues, and cells in vivo have not been previously described.
  • the many drawbacks of existing synthetic RNA technologies and methods for delivery of nucleic acids make them undesirable for therapeutic or cosmetic use.
  • RNA-based reprogramming methods have been described (see, e.g., Angel. MIT Thesis. 2008. 1-56; Angel et al. PLoS ONE. 2010. 5, 107; Warren et al. Cell Stem Cell. 2010. 7, 618-630; Angel.
  • RNA-based reprogramming methods are slow, unreliable, and inefficient when performed on adult cells, require many transfections (resulting in significant expense and opportunity for error), can reprogram only a limited number of cell types, can reprogram cells to only a limited number of cell types, require the use of immunosuppressants, and require the use of multiple human-derived components, including blood-derived HSA and human fibroblast feeders.
  • the many drawbacks of previously disclosed RNA-based reprogramming methods make them undesirable for research, therapeutic or cosmetic use.
  • DNA-binding domains that can recognize specific DNA sequences, for example, zinc fingers (ZFs) and transcription activator-like effectors (TALEs). Fusion proteins containing one or more of these DNA-binding domains and the cleavage domain of FokI endonuclease can be used to create a double-strand break in a desired region of DNA in a cell (see, e.g., US Patent Appl. Pub. No. US 2012/0064620, US Patent Appl. Pub. No. US 2011/0239315, U.S. Pat. No. 8,470,973, US Patent Appl. Pub. No. US 2013/0217119, U.S. Pat. No. 8,420,782, US Patent Appl. Pub.
  • gene-editing proteins include clustered regularly interspaced short palindromic repeat (CRISPR)-associated proteins.
  • CRISPR clustered regularly interspaced short palindromic repeat
  • the present invention provides, in part, compositions, methods, articles, and devices for delivering nucleic acids to cells, tissues, organs, and patients, methods for inducing cells to express proteins, methods, articles, and devices for producing these compositions, methods, articles, and devices, and compositions and articles, including cells, organisms, cosmetics and therapeutics, produced using these compositions, methods, articles, and devices.
  • certain embodiments of the present invention provide small doses of nucleic acids to achieve significant and lasting protein expression in humans.
  • the present invention is based on the surprising discovery of safe and effective doses and administration parameters for nucleic acid drugs in human subjects.
  • a method for delivering a nucleic acid drug comprising administering an effective dose of a nucleic acid drug to a human subject in need thereof.
  • the nucleic acid drug comprises a RNA comprising one or more non-canonical nucleotides (a/k/a “modified RNA”).
  • the effective dose is an amount sufficient to substantially increase an amount of a protein encoded by the nucleic acid drug in the human subject and/or substantially avoid an immune reaction in a human subject, wherein the immune reaction is optionally mediated by the innate immune system.
  • a method for expressing a protein of interest in a population of cells in a mammalian subject comprising administering a non-viral transfection composition comprising an effective dose of a RNA encoding the protein of interest to said cells, where the transfection composition is administered in an amount that allows for expression of said protein in said cells for at least about six hours to about five days without substantial cellular toxicity.
  • the RNA contains one or more non-canonical nucleotides that avoid substantial cellular toxicity.
  • the effective dose is about 100 ng to about 2000 ng (e.g., about, or no more than about, 100 ng, or 200 ng, or 300 ng, or 400 ng, or 500 ng, or 600 ng, or 700 ng, or 800 ng, or 900 ng, or 1000 ng, or 1100 ng, or 1200 ng, or 1300 ng, or 1400 ng, or 1500 ng, or 1600 ng, or 1700 ng, or 1800 ng, or 1900 ng, or 2000 ng). In other embodiments, the effective dose is less than about 100 ng.
  • the effective dose is about 10 ng to about 100 ng (e.g., about, or no more than about, 10 ng, or 20 ng, or 30 ng, or 40 ng, or 50 ng, or 60 ng, or 70 ng, or 80 ng, or 90 ng, or 100 ng).
  • the effective dose is about 1.4 ng/kg to about 30 ng/kg (e.g., about, or no more than about, 1.4 ng/kg, or 2.5 ng/kg, or 5 ng/kg, or 10 ng/kg, or 15 ng/kg, or 20 ng/kg, or 25 ng/kg, or 30 ng/kg. In other embodiments, the effective dose is less than about 1.5 ng/kg.
  • the effective dose is about 0.14 ng/kg to about 1.4 ng/kg (e.g., about, or no more than about, 0.14 ng/kg, or 0.25 ng/kg, or 0.5 ng/kg, or 0.75 ng/kg, or 1 ng/kg, or 1.25 ng/kg, or 1.4 ng/kg).
  • the effective dose is about 350 ng/cm 2 to about 7000 ng/cm 2 (e.g., about, or no more than about, 350 ng/cm 2 , or 500 ng/cm 2 , or 750 ng/cm 2 , or 1000 ng/cm 2 , or 2000 ng/cm 2 , or 3000 ng/cm 2 , or 4000 ng/cm 2 , or 5000 ng/cm 2 , or 6000 ng/cm 2 , or 7000 ng/cm 2 ). In other embodiments, the effective dose is less than about 350 ng/cm 2 .
  • the effective dose is about 35 ng/cm 2 to about 350 ng/cm 2 (e.g., about, or no more than about, 35 ng/cm 2 , or 50 ng/cm 2 , or 75 ng/cm 2 , or 100 ng/cm 2 , or 150 ng/cm 2 , or 200 ng/cm 2 , or 250 ng/cm 2 , or 300 ng/cm 2 , or 350 ng/cm 2 .
  • ng/cm 2 e.g., about, or no more than about, 35 ng/cm 2 , or 50 ng/cm 2 , or 75 ng/cm 2 , or 100 ng/cm 2 , or 150 ng/cm 2 , or 200 ng/cm 2 , or 250 ng/cm 2 , or 300 ng/cm 2 , or 350 ng/cm 2 .
  • the effective dose is about 0.28 picomoles to about 5.7 picomoles (e.g., about, or no more than about, 0.28 picomoles, or 0.5 picomoles, or 0.75 picomoles, or 1 picomole, or 2 picomoles, or 3 picomoles, or 4 picomoles, or 5 picomoles, or 5.7 picomoles). In other embodiments, the effective dose is less than about 0.28 picomoles.
  • the effective dose is about 0.028 picomoles to about 0.28 picomoles (e.g., about, or no more than about, 0.028 picomoles, or 0.05 picomoles, or 0.075 picomoles, or 0.1 picomoles, or 0.15 picomoles, or 0.2 picomoles, or 0.25 picomoles, or 0.28 picomoles).
  • the effective dose is about 0.004 picomoles/kg to about 0.082 picomoles/kg (e.g., about, or no more than about, 0.004 picomoles/kg, or 0.01 picomoles/kg, or 0.02 picomoles/kg, or 0.03 picomoles/kg, or 0.04 picomoles/kg, or 0.05 picomoles/kg, or 0.06 picomoles/kg, or 0.07 picomoles/kg, or 0.08 picomoles/kg, or 0.082 picomoles/kg). In other embodiments, the effective dose is less than about 0.004 picomoles/kg.
  • the effective dose is about 0.0004 picomoles/kg to about 0.004 picomoles/kg (e.g., about, or no more than about, 0.0004 picomoles/kg, or 0.001 picomoles/kg, or 0.002 picomoles/kg, or 0.003 picomoles/kg, or 0.004 picomoles/kg).
  • the effective dose is about 1 picomole/cm 2 to about 20 picomoles/cm 2 (e.g., about, or no more than about, 1 picomole/cm 2 , or 2 picomoles/cm 2 , or 3 picomoles/cm 2 , or 4 picomoles/cm 2 , or 5 picomoles/cm 2 , or 6 picomoles/cm 2 , or 7 picomoles/cm 2 , or 8 picomoles/cm 2 , or 9 picomoles/cm 2 , or 10 picomoles/cm 2 , or 12 picomoles/cm 2 , or 14 picomoles/cm 2 , or 16 picomoles/cm 2 , or 18 picomoles/cm 2 , or 20 picomoles/cm 2 ).
  • the effective dose is less than about 1 picomole/cm 2 .
  • the effective dose is about 0.1 picomoles/cm 2 to about 1 picomole/cm 2 (e.g., about, or no more than about, 0.1 picomoles/cm 2 , or 0.2 picomoles/cm 2 , or 0.3 picomoles/cm 2 , or 0.4 picomoles/cm 2 , or 0.5 picomoles/cm 2 , or 0.6 picomoles/cm 2 , or 0.7 picomoles/cm 2 , or 0.8 picomoles/cm 2 , or 0.9 picomoles/cm 2 , or 1 picomole/cm 2 ).
  • the nucleic acid drug is administered in a pharmaceutically acceptable formulation, including a formulation suitable for one or more of injection (e.g. subcutaneous injection, intradermal injection (including to the dermis or epidermis), subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection) and topical administration and/or for administration to the integumentary system (e.g.
  • injection e.g. subcutaneous injection, intradermal injection (including to the dermis or epidermis), subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection
  • topical administration and/or for administration to the integumentary system e.g.
  • the epidermis optionally selected from the stratum corneum, stratum lucidum, stratum granulosum, stratum spinosum, and stratum germinativum
  • basement membrane optionally selected from the papillary region and the reticular region
  • subcutis optionally selected from the papillary region and the reticular region
  • conjunctiva e.g., to one or more of the cornea, sclera, iris, lens, corneal limbus, optic nerve, choroid, ciliary body, anterior segment, anterior chamber, and retina.
  • the nucleic acid drug is formulated with one or more lipids to enhance uptake of the nucleic acid drug by cells, the lipids optionally selected from Table 1.
  • the nucleic acid drug is formulated with one or more nanoparticles, optionally lipid or polymeric nanoparticles, to enhance uptake of the nucleic acid drug by cells, to enhance duration of protein expression, or to otherwise enhance the safety and/or efficacy of the nucleic acid drug.
  • the nucleic acid drug is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose is administered to a surface area of about 4 mm 2 to about 1000 mm 2 (e.g. about, or no more than about, 4 mm 2 , or 5 mm 2 , or 10 mm 2 , or 25 mm 2 , or 50 mm 2 , or 75 mm 2 , or 100 mm 2 , or 125 mm 2 , or 150 mm 2 , or 200 mm 2 , or 500 mm 2 , or 1000 mm 2 ).
  • mm 2 e.g. about, or no more than about, 4 mm 2 , or 5 mm 2 , or 10 mm 2 , or 25 mm 2 , or 50 mm 2 , or 75 mm 2 , or 100 mm 2 , or 125 mm 2 , or 150 mm 2 , or 200 mm 2 , or 500 mm 2 , or 1000
  • the nucleic acid drug is administered in a treatment regimen, optionally with an additional agent or adjuvant therapy described herein, and the administration is about weekly to about once every 24 weeks (e.g. about, or not more than about, weekly, or once every 2 weeks, or once every 3 weeks, or once every 4 weeks, or once every 5 weeks, or once every 6 weeks, or once every 7 weeks, or once every 8 weeks, or once every 9 weeks, or once every 9 weeks, or once every 9 weeks, or once every 9 weeks, or once every 10 weeks, or once every 11 weeks, or once every 12 weeks, or once every 13 weeks, or once every 14 weeks, or once every 15 weeks, or once every 20 weeks, or once every 24 weeks).
  • once every 24 weeks e.g. about, or not more than about, weekly, or once every 2 weeks, or once every 3 weeks, or once every 4 weeks, or once every 5 weeks, or once every 6 weeks, or once every 7 weeks, or once every 8 weeks, or once every 9 weeks, or once every 9 weeks, or once every 9 weeks, or once every
  • the nucleic acid drug is administered in a treatment regimen, optionally with an additional agent or adjuvant therapy described herein, and the administration is about daily to about weekly (e.g., about, or not more than about, daily, or once every 2 days, or once every 3 days, or once every 4 days, or once every 5 days, or once every 6 days, or weekly).
  • the nucleic acid drug comprises RNA comprising one or more non-canonical nucleotides, optionally having one or more substitutions at the 2C and/or 4C and/or 5C positions for a pyrimidine or the 6C and/or 7N and/or 8C positions for a purine.
  • the non-canonical nucleotide is one or more of the non-canonical nucleotides described herein, including, for example, 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methyluridine, 5-methylpseudouridine, 5-hydroxymethyluridine, 5-hydroxymethylpseudouridine, 5-carboxyuridine, 5-carboxypseudouridine, 5-formyluridine, 5-formylpseudouridine, 5-methoxyuridine, and 5-methoxypseudouridine.
  • RNA comprising one or more non-canonical nucleotides may have one or more of a 5′-UTR comprising a Kozak consensus sequence, a 5′-UTR or 3′-UTR comprising a sequence that increases RNA stability in vivo (e.g. an alpha-globin or beta-globin 5′-UTR or an alpha-globin or beta-globin 3′-UTR), and a 3′ poly(A) tail from about 20 nucleotides to about 250 nucleotides in length (e.g.
  • aspects of the methods described herein find use in various medical treatments, including, by way of illustration, treating a disease, disorder and/or condition of the integumentary system or altering, modifying and/or changing the integumentary system (e.g. cosmetically).
  • kits suitable for use in human therapy comprising the nucleic acid drug described herein in a unit dosage form of about 10 ng to about 2000 ng (e.g. about, or no more than about, 10 ng, or 20 ng, or 50 ng, or 100 ng, or 200 ng, or 300 ng, or 400 ng, or 500 ng, or 600 ng, or 700 ng, or 800 ng, or 900 ng, or 1000 ng, or 1100 ng, or 1200 ng, or 1300 ng, or 1400 ng, or 1500 ng, or 1600 ng, or 1700 ng, or 1800 ng, or 1900 ng, or 2000 ng) and an injection needle.
  • 10 ng to about 2000 ng e.g. about, or no more than about, 10 ng, or 20 ng, or 50 ng, or 100 ng, or 200 ng, or 300 ng, or 400 ng, or 500 ng, or 600 ng, or 700 ng, or 800 ng, or 900 ng
  • the present invention provides a pharmaceutical formulation comprising the nucleic acid drug described herein and one or more of the vehicles (a/k/a “transfection reagents”, e.g., lipids) described herein, the formulation optionally being suitable for one or more of subcutaneous injection, intradermal injection, subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection, and topical administration.
  • the vehicles a/k/a “transfection reagents”, e.g., lipids
  • nucleic acid delivery patches are provided.
  • devices for delivering nucleic acids using electric fields are provided.
  • Other aspects pertain to methods and compositions for delivery of nucleic acids to the skin.
  • Still further aspects pertain to methods and compositions for expression of proteins in the skin.
  • the invention provides methods and compositions for treating diseases and conditions in humans, including, but not limited to, prophylactic treatments, treatments for rare diseases, including, but not limited to, dermatologic rare diseases, and treatments for use in medical dermatology and aesthetic medicine.
  • the invention provides cosmetics comprising nucleic acids.
  • Still further aspects relate to methods and compositions for altering pigmentation, for example, for the treatment of pigmentation disorders.
  • Still further aspects relate to methods and compositions for enhancing healing, including, but not limited to, healing in response to a wound or surgery.
  • the compositions of the present invention may alter, modify and/or change the appearance of a member of the integumenary system of a subject such as, but not limited, to skin, hair and nails. Such alteration, modification and/or change may be in the context of treatment methods and/or therapeutic uses as described herein including, by way of non-limiting example, dermatological treatments and cosmetics procedures.
  • the present invention relates to the targeting of various therapeutic proteins that are not limited to dermatological applications.
  • the present compositions and methods find use in methods of treatment that are mediated by the increased expression of, for example, various soluble proteins as illustrated herein.
  • the nucleic acid drug encodes and/or increases the expression of one or more of a circulating protein, an extracellular matrix protein, an engineered protein, a gene-editing protein, a protein or peptide hormone, an enzyme, erythropoietin, darbepoetin alfa, NOVEPOETIN, elastin, collagen, an antibody or antibody fragment (e.g., a neutralizing antibody or antibody fragment), an intracellular protein, telomerase reverse transcriptase, a membrane protein, a fusion protein, a receptor, a ligand binding domain, a protein inhibitor, or a biologically active fragment, analogue or variant thereof.
  • a circulating protein an extracellular matrix protein
  • an engineered protein e.g., a gene-editing protein, a protein or peptide hormone, an enzyme, erythropoietin, darbepoetin alfa, NOVEPOETIN, elastin, collagen, an antibody or antibody fragment (e.
  • administration of the nucleic acid drug results in one or more of an increase in hematocrit, an increase in tissue elasticity, an increase in tissue strength, and increase in skin hydration and/or water retention, hair growth, fat reduction, an insertion, deletion or mutation in DNA, conversion of a prodrug to an active drug, a decrease in tumor size and/or number, a decrease in plaque size and/or number, an increase in vascularization, a decrease in vascularization, an increase in visual acuity, a decrease in pain, an increase in cardiac output (e.g., ejection fraction and stroke volume), a decrease in abnormal heart rhythm, a decrease in fibrosis, a decrease in one or more adverse neurological symptoms, conditions, or disorders (e.g., depression, dysregulation of appetite, polyphagia, anorexia, dementia, headache, fatigue, numbness, tremors, and dizziness), a decrease in erectile dysfunction, an increase in vitality, an increase in pulmonary function, an increase in
  • RNA molecules with low toxicity and high translation efficiency are provided.
  • a cell-culture medium for high-efficiency in vivo transfection, reprogramming, and gene editing of cells is provided.
  • Other aspects pertain to methods for producing RNA molecules encoding reprogramming proteins.
  • Still further aspects pertain to methods for producing RNA molecules encoding gene-editing proteins.
  • the invention provides high-efficiency gene-editing proteins comprising engineered nuclease cleavage domains. In another aspect, the invention provides high-fidelity gene-editing proteins comprising engineered nuclease cleavage domains. Other aspects relate to high-efficiency gene-editing proteins comprising engineered DNA-binding domains. Still further aspects pertain to high-fidelity gene-editing proteins comprising engineered DNA-binding domains. Still further aspects relate to gene-editing proteins comprising engineered repeat sequences. Some aspects relate to methods for altering the DNA sequence of a cell by transfecting the cell with or inducing the cell to express a gene-editing protein. Other aspects relate to methods for altering the DNA sequence of a cell that is present in an in vitro culture. Still further aspects relate to methods for altering the DNA sequence of a cell that is present in vivo.
  • the invention provides methods for treating cancer comprising administering to a patient a therapeutically effective amount of a gene-editing protein or a nucleic-acid encoding a gene-editing protein.
  • the gene-editing protein is capable of altering the DNA sequence of a cancer associated gene.
  • the cancer-associated gene is the BIRC5 gene.
  • nucleic acids and/or cells relate to therapeutics comprising nucleic acids and/or cells and methods of using therapeutics comprising nucleic acids and/or cells for the treatment of, for example, type 1 diabetes, heart disease, including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, cystic fibrosis, sickle-cell anemia, thalassemia, Fanconi anemia, severe combined immunodeficiency, hereditary sensory neuropathy, xeroderma pigmentosum, Huntington's disease, muscular dystrophy, amyotrophic lateral sclerosis, Alzheimer's disease, cancer, and infectious diseases including hepatitis and HIV/AIDS.
  • the nucleic acids comprise RNA.
  • the RNA comprises one or more non-canonical nucleotides.
  • the nucleic acids are delivered to cells using a virus.
  • the virus is a replication-competent virus. In other aspects, the virus is a replication-incompetent virus.
  • FIG. 1 depicts primary adult human dermal fibroblasts transfected with RNA encoding green fluorescent protein (“GFP”) and comprising the indicated nucleotides.
  • GFP green fluorescent protein
  • FIG. 2 depicts the result of a gene-expression analysis of the primary adult human dermal fibroblasts of FIG. 1 using a one-step real-time RT-PCR and primers designed to amplify human interferon beta mRNA. Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 3 depicts the results of a gene-expression analysis of cells transfected with RNA comprising the indicated nucleotides, conducted as in FIG. 2 . Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 4 depicts the results of a gene-expression analysis of cells transfected with RNA comprising the indicated nucleotides, conducted as in FIG. 2 , and using primers designed to amplify the indicated transcripts. Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 5 depicts the results of a gene-expression analysis of primary human epidermal keratinocytes transfected with RNA encoding NOVEPOETIN, and comprising the indicated nucleotides, conducted as in FIG. 2 . Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 6 depicts intradermal injection of a solution comprising RNA encoding GFP into the ventral forearm of a healthy, 33 year-old, 70 kg, male human subject.
  • FIG. 7 depicts a region of the ventral forearm of the subject shown in FIG. 6 after treatment with RNA comprising 5-methoxyuridine and encoding GFP (injection sites 1-3) or COL7 (injection site 4). The image was taken immediately following the final injection.
  • FIG. 8 depicts the region of FIG. 7 , 24 hours after injection.
  • FIG. 9 depicts the results of fluorescent imaging of the region of FIG. 7 , using the indicated fluorescent channels. The dose at each injection site is also indicated. Images were taken 24 hours after injection.
  • FIG. 10 depicts the results of fluorescent imaging of the region of FIG. 7 , using the FITC fluorescent channel. The dose at each injection site is indicated. Images were taken 48 hours after injection.
  • FIG. 11 depicts the results of quantitative fluorescent imaging of the region of FIG. 7 , using the FITC fluorescent channel.
  • the horizontal axis indicates time after injection.
  • FIG. 12 depicts the results of fluorescent imaging of an independent experiment in which a region of the ventral forearm of as the subject shown in FIG. 6 treated with RNA comprising 5-methoxyuridine and encoding GFP. The image was taken 24 hours after injection.
  • FIG. 13 depicts the results of an ELISA designed to detect darbepoetin alfa in culture media of primary human epidermal keratinocytes transfected with RNA comprising the indicated nucleotides and encoding NOVEPOETIN.
  • FIG. 14 depicts the results of an ELISA designed to detect darbepoetin alfa in culture media of primary human epidermal keratinocytes transfected with RNA comprising the indicated nucleotides and encoding NOVEPOETIN.
  • FIG. 15 depicts the results of an ELISA designed to detect darbepoetin alfa in culture media of primary human epidermal keratinocytes transfected with RNA comprising the indicated nucleotides and encoding NOVEPOETIN.
  • FIG. 16 depicts primary human dermal fibroblasts transfected with RNA comprising 5-methoxyuridine and encoding hTERT. Cells were fixed and stained using an antibody targeting hTERT 24 hours after transfection.
  • FIG. 17 depicts primary adult human dermal fibroblasts transfected with RNA encoding green fluorescent protein (“GFP”), prepared and stored as indicated.
  • GFP green fluorescent protein
  • FIG. 18 depicts single administration of NOVECRIT induced a rapid increase and sustained level of NOVEPOIETIN in serum.
  • the Y axis shows concentration of NOVEPOIETIN protein (mU/mL).
  • FIG. 19 depicts a single administration of NOVECRIT stimulated erythropoiesis, yielding elevated hematocrit for at least 14 days.
  • the left panel shows % hematocrit on the Y axis, while the right panel shows % reticulocytes.
  • FIG. 20 depicts a table summarizing TNF ⁇ , IL-6, and IFN ⁇ cytokine levels in plasma samples collected from a maximum tolerated dose of NOVECRIT in male Sprague Dawley rats study of Example 35.
  • FIG. 21 depicts a SURVEYOR assay using the DNA of primary adult human dermal fibroblasts transfected with RNA TALENs targeting the sequence TGAGCAGAAGTGGCTCAGTG (SEQ ID NO:467) and TGGCTGTACAGCTACACCCC (SEQ ID NO: 468), located within the COL7A1 gene.
  • the bands present in the +RNA lane indicate editing of a region of the gene that is frequently involved in dystrophic epidermolysis bullosa.
  • FIG. 22 depicts a SURVEYOR assay using the DNA of primary adult human dermal fibroblasts transfected with RNA TALENs targeting the sequence TTCCACTCCTGCAGGGCCCC (SEQ ID NO:469) and TCGCCCTTCAGCCCGCGTTC (SEQ ID NO:470), located within the COL7A1 gene.
  • the bands present in the +RNA lane indicate editing of a region of the gene that is frequently involved in dystrophic epidermolysis bullosa.
  • FIG. 23 shows the immunogenicity of various synthetic RNA constructs in the context of a gene-editing (i.e. unmodified nucleotides “A,G,U,C”; pseudouridine only “psU”; 5-methylcytidine only “5mC”; both pseudouridine and 5-methylcytidine “psU+5mC”; and a negative control “neg”).
  • a gene-editing i.e. unmodified nucleotides “A,G,U,C”; pseudouridine only “psU”; 5-methylcytidine only “5mC”; both pseudouridine and 5-methylcytidine “psU+5mC”; and a negative control “neg”).
  • FIG. 24 shows the gene-editing activity in cells transfected with various synthetic RNA constructs (i.e. unmodified nucleotides “A,G,U,C”; psuedouridine only “psU”; 5-methylcytidine only “5mC”; both psuedouridine and 5-methylcytidine “psU+5mC”; and a negative control “neg”).
  • synthetic RNA constructs i.e. unmodified nucleotides “A,G,U,C”; psuedouridine only “psU”; 5-methylcytidine only “5mC”; both psuedouridine and 5-methylcytidine “psU+5mC”; and a negative control “neg”).
  • FIG. 25 depicts gene editing of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and a single-stranded DNA repair template (“RT”) of the indicated length.
  • RT single-stranded DNA repair template
  • * indicates successful gene editing.
  • FIG. 26 depicts gene editing of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and an 80 nt single-stranded DNA repair template (“RT”) at the indicated ratios of RNA to repair template.
  • RT single-stranded DNA repair template
  • * indicates successful gene editing.
  • FIG. 27 depicts gene correction of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and a single-stranded DNA repair template (“RT”) of the indicated length.
  • RT single-stranded DNA repair template
  • * indicates successful gene correction.
  • FIG. 28 depicts gene correction of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and an 80 nt single-stranded DNA repair template (“RT”) at the indicated ratios of RNA to repair template.
  • RT single-stranded DNA repair template
  • * indicates successful gene correction.
  • FIG. 29 depicts gene editing (“T7E1”) and correction (“Digestion”) of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and an 80 nt single-stranded DNA repair template (“RT”).
  • RT 80 nt single-stranded DNA repair template
  • * indicates successful gene editing (“T7E1”) and correction (“Digestion”).
  • FIG. 30 depicts the amount of BMP7 protein secreted by primary human dermal fibroblasts and primary human epidermal keratinocytes transfected with RNA encoding the indicated BMP7 variant, measured by ELISA.
  • Asterisks (“*”) indicate saturation of the ELISA assay.
  • FIG. 31 depicts the amount of parathyroid hormone secreted by primary human epidermal keratinocytes transfected with RNA encoding PTH, measured by ELISA.
  • FIG. 32 demonstrates that repeated administration of NOVECRIT stimulated erythropoiesis, yielding elevated hematocrit for at least 14 days.
  • the order of histograms from left to right is: Group 1, Group 2, Group 3, Group 4, and Group 5.
  • FIG. 33 provides an illustrative experimental design for studying the effects of RNAs encoding BMP7 variants for the prevention and treatment of diabetic nephropathy.
  • FIG. 35 depicts the urine volume, urine albumin, and urine creatinine levels in rats treated with RNAs encoding BMP7 variants as described in Example 42.
  • the order of histograms from left to right is: urine volume, urine creatinine, and urine albumin.
  • FIG. 36 depicts the effect of RNAs encoding BMP7 variants in treating diabetic nephropathy as described in Example 42.
  • the order of histograms from left to right is: Group 6—vehicle and Group 7—FTB-2 (BMP7 variant A).
  • FIG. 37 panels A-I, depict the results of gene-expression analysis of human epidermal keratinocytes transfected with RNA encoding BDNF, BMP-2, BMP-6, IL-2, IL-6, IL-15, IL-22, LIF or FGF-21.
  • FIG. 38 depicts the results of gene-expression analysis of human epidermal keratinocytes transfected with RNA encoding IL-15 or IL-15 and IL-15RA.
  • FIG. 39 depicts the serum levels of FGF21, IL15, IL6, IL22, and Novepoietin following a single intradermal injection of various RNAs encoding these proteins as described in Example 45. Three rats were analyzed for each RNA tested.
  • the present invention is based, in part, on the discovery of a safe and effective dosing strategy for nucleic acid drugs, including RNA, such as RNA comprising non-canonical (or “modified”) nucleotides, in humans.
  • RNA such as RNA comprising non-canonical (or “modified”) nucleotides
  • the inventors believe this to be the first report of safe and effective dosing of RNA molecules, including those comprising non-canonical nucleotides, in humans.
  • very large doses of RNA molecules are needed for mammalian dosing, and minimal therapeutic effect is achieved despite high dosing (see, e.g. US Patent Publication No. 2013/0245103)
  • the present inventors have surprisingly managed to dose synthetic RNA in a human and achieve significant target protein expression with minimal immunological or other side effects.
  • the present invention provides improved doses, formulations, administration and methods of use of nucleic acid drugs, which include RNA, which may contain non-canonical nucleotides (e.g. a residue other than adenine, guanine, thymine, uracil, and cytosine or the standard nucleoside, nucleotide, deoxynucleoside or deoxynucleotide derivatives thereof).
  • RNA comprising non-canonical nucleotides leads to the expression of a protein encoded by the RNA, the protein often being one of therapeutic benefit (sometimes called the “target” or “protein of interest”). Further, this expression of therapeutic protein is achieved with minimal or negligible toxicity.
  • nucleic acid drugs include a dsDNA molecule, a ssDNA molecule, a RNA molecule, a dsRNA molecule, a ssRNA molecule, a plasmid, an oligonucleotide, a synthetic RNA molecule, a miRNA molecule, an mRNA molecule, and an siRNA molecule.
  • the RNA comprises non-canonical nucleotides.
  • a method for delivering a nucleic acid drug comprising administering an effective dose of a nucleic acid drug to a human subject in need thereof, wherein the nucleic acid drug comprises a synthetic RNA.
  • the effective dose is an amount sufficient to substantially increase an amount of a protein encoded by the nucleic acid drug in the human subject.
  • the nucleic acid drug is a synthetic RNA comprising one or more modified nucleotides
  • the nucleic acid drug may result in higher protein expression than levels obtainable with a nucleic acid drug that does not comprise one or more modified nucleotides (e.g., RNA comprising the canonical nucleotides A, G, U, and C).
  • the nucleic acid drug results in about a 2-fold, or about a 3-fold, or about a 4-fold, or about a 5-fold, or about a 10-fold, or about a 15-fold, or about a 20-fold, or about a 25-fold, or about a 30-fold, or about a 35-fold, or about a 40-fold, or about a 45-fold, or about a 50-fold, or about a 100-fold increase in protein expression as compared to levels obtainable with a nucleic acid drug that does not comprise one or more modified nucleotides.
  • the nucleic acid drug provides a sustained therapeutic effect that is optionally mediated by a sustained expression of target protein.
  • the therapeutic effect is present for over about 1 day, or over about 2 days, or over about 3 days, or over about 4 days, or over about 5 days, or over about 6 days, or over about 7 days, or over about 8 days, or over about 9 days, or over about 10 days, or over about 14 days after administration.
  • this sustained effect obviates the need for, or reduces the amount of, maintenance doses.
  • the nucleic acid drug provides a sustained target protein level.
  • the target protein is present (e.g. in measurable amounts, e.g. in the serum of a patient to whom the nucleic acid drug has been administered) for over about 1 day, or over about 2 days, or over about 3 days, or over about 4 days, or over about 5 days, or over about 6 days, or over about 7 days, or over about 8 days, or over about 9 days, or over about 10 days, or over about 14 days after administration.
  • this sustained effect obviates the need for, or reduces the amount of, maintenance doses.
  • the nucleic acid drug provides therapeutic action without sustained presence of the nucleic acid drug itself.
  • the nucleic acid drug is rapidly metabolized, for instance, within about 6 hours, or about 12 hours, or about 18 hours, or about 24 hours, or about 2 days, or about 3 days, or about 4 days, or about 5 days, or about 1 week from administration.
  • the effective dose is an amount that substantially avoids cell toxicity in vivo. In various embodiments, the effective dose is an amount that substantially avoids an immune reaction in a human subject.
  • the immune reaction may be an immune response mediated by the innate immune system. Immune response can be monitored using markers known in the art (e.g. cytokines, interferons, TLRs).
  • the effective dose obviates the need for treatment of the human subject with immune suppressants agents (e.g. B18R) used to moderate the residual toxicity. Accordingly, in some embodiments, the present methods allow for dosing that provides increased protein expression and reduces toxicity.
  • the immune response is reduced by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, about 99.9%, or greater than about 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid.
  • upregulation of one or more immune response markers is reduced by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, about 99.9%, or greater than about 99.9% as compared to the upregulation of the one or more immune response markers induced by a corresponding unmodified nucleic acid.
  • the immune response marker comprises an mRNA or protein product of an interferon gene, including an interferon alpha gene, IFNB1, TLR3, RARRES3, EIF2AK2, STAT1, STAT2, IFIT1, IFIT2, IFIT3, IFIT5, OAS1, OAS2, OAS3, OASL, ISG20 or a fragment, variant, analogue, or family-member thereof.
  • the immune response marker comprises an mRNA or protein product of an TNF gene, including an TNF alpha gene, TNFRSF1A; TNFRSF1B; LTBR; TNFRSF4; CD40; FAS; TNFRSF6B; CD27; TNFRSF8; TNFRSF9; TNFRSF10A; TNFRSF10B; TNFRSF10C; TNFRSF10D; TNFRSF11A; TNFRSF11B; TNFRSF12A; TNFRSF13B; TNFRSF13C; TNFRSF14; NGFR; TNFRSF17; TNFRSF18; TNFRSF19; TNFRSF21; TNFRSF25; and EDA2R or a fragment, variant, analogue, or family-member thereof.
  • TNF gene including an TNF alpha gene, TNFRSF1A; TNFRSF1B; LTBR; TNFRSF4; CD40; FAS; TNFRSF6B; CD
  • the immune response marker comprises an mRNA or protein product of an interleukin gene, including an IL-6 gene, IL-1; IL-2; IL-3; IL-4; IL-5; IL-6; IL-7; IL-8 or CXCL8; IL-9; IL-10; IL-11; IL-12; IL-13; IL-14; IL-15; IL-16; IL-17; IL-18; IL-19; IL-20; IL-21; IL-22; IL-23; IL-24; IL-25; IL-26; IL-27; IL-28; IL-29; IL-30; IL-31; IL-32; IL-33; IL-35; IL-36 or a fragment, variant, analogue, or family-member thereof.
  • an interleukin gene including an IL-6 gene, IL-1; IL-2; IL-3; IL-4; IL-5; IL-6; IL-7; IL-8 or CXCL8; IL-9;
  • cell death is about 10%, about 25%, about 50%, about 75%, about 85%, about 90%, about 95%, or over about 95% less than the cell death observed with a corresponding unmodified nucleic acid. Moreover, cell death may affect fewer than about 50%, about 40%, about 30%, about 20%, about 10%, about 5%, about 1%, about 0.1%, about 0.01% or fewer than about 0.01% of cells contacted with the modified nucleic acids.
  • a method for expressing a protein of interest in a population of cells in a mammalian subject comprising administering a non-viral transfection composition comprising an effective dose of a RNA encoding the protein of interest to said cells, the RNA containing one or more non-canonical nucleotides that avoid substantial cellular toxicity, where the transfection composition is administered in an amount that allows for expression of said protein in said cells for at least about five days (e.g. about 5, or about 6, or about 7, about 8, or about 9, or about 10, or about 14 days) without substantial cellular toxicity.
  • a method for expressing a protein of interest in a population of cells in a mammalian subject comprising administering a non-viral transfection composition comprising an effective dose of a RNA encoding the protein of interest to said cells, the RNA containing one or more non-canonical nucleotides that avoid substantial cellular toxicity, where the transfection composition is administered in an amount that allows for expression of said protein in said cells for at least about six hours (e.g. about six hours, or about 12 hours, or about 1 day, or about 2 days, or about 3 days, or about 4 days, or about 5 days) without substantial cellular toxicity.
  • the effective dose of the nucleic acid drug, including synthetic RNA is about 100 ng to about 2000 ng, or about 200 ng to about 1900 ng, or about 300 ng to about 1800 ng, or about 400 ng to about 1700 ng, or about 500 ng to about 1600 ng, or about 600 ng to about 1500 ng, or about 700 ng to about 1400 ng, or about 800 ng to about 1300 ng, or about 900 ng to about 1200 ng, or about 1000 ng to about 1100 ng, or about 500 ng to about 2000 ng, or about 500 ng to about 1500 ng, or about 500 ng to about 1000 ng, or about 1000 ng to about 1500 ng, or about 1000 ng to about 2000 ng, or about 1500 ng to about 2000 ng, or about 100 ng to about 500 ng, or about 200 ng to about 400 ng, or about 10 ng to about 100 ng, or about 20 ng to about 90 ng, or about 30 ng to about 80
  • the effective dose of the nucleic acid drug, including synthetic RNA is no more than about 50 ng, or about 100 ng, or about 200 ng, or about 300 ng, or about 400 ng, or about 500 ng, or about 600 ng, or about 700 ng, or about 800 ng, or about 900 ng, or about 1000 ng, or about 1100 ng, or about 1200 ng, or about 1300 ng, or about 1400 ng, or about 1500 ng, or about 1600 ng, or about 1700 ng, or about 1800 ng, or about 1900 ng, or about 2000 ng.
  • the effective dose of the nucleic acid drug, including synthetic RNA is about 50 ng, or about 100 ng, or about 200 ng, or about 300 ng, or about 400 ng, or about 500 ng, or about 600 ng, or about 700 ng, or about 800 ng, or about 900 ng, or about 1000 ng, or about 1100 ng, or about 1200 ng, or about 1300 ng, or about 1400 ng, or about 1500 ng, or about 1600 ng, or about 1700 ng, or about 1800 ng, or about 1900 ng, or about 2000 ng.
  • the effective dose of the nucleic acid drug, including synthetic RNA is about 0.028 pmol, or about 0.05 pmol, or about 0.1 pmol, or about 0.2 pmol, or about 0.3 pmol, or about 0.4 pmol, or about 0.5 pmol, or about 0.6 pmol, or about 0.7 pmol, or about 0.8 pmol, or about 0.9 pmol, or about 1.0 pmol, or about 1.2 pmol, or about 1.4 pmol, or about 1.6 pmol, or about 1.8 pmol, or about 2.0 pmol, or about 2.2 pmol, or about 2.4 pmol, or about 2.6 pmol, or about 2.8 pmol, or about 3.0 pmol, or about 3.2 pmol, or about 3.4 pmol, or about 3.6 pmol, or about 3.8 pmol, or about 4.0 pmol, or about 4.2 pmol, or about 4.4 pmol, or about
  • the nucleic acid drug including synthetic RNA, is administered at a concentration of about 0.1 nM, or about 0.25 nM, or about 0.5 nM, or about 0.75 nM, or about 1 nM, or about 2.5 nM, or about 5 nM, or about 7.5 nM, or about 10 nM, or about 20 nM, or about 30 nM, or about 40 nM, or about 50 nM, or about 60 nM, or about 70 nM, or about 80 nM, or about 90 nM, or about 100 nM, or about 110 nM, or about 120 nM, or about 150 nM, or about 175 nM, or about 200 nM.
  • the effective dose of the nucleic acid drug is about 350 ng/cm 2 , or about 500 ng/cm 2 , or about 750 ng/cm 2 , or about 1000 ng/cm 2 , or about 2000 ng/cm 2 , or about 3000 ng/cm 2 , or about 4000 ng/cm 2 , or about 5000 ng/cm 2 , or about 6000 ng/cm 2 , or about 7000 ng/cm 2 . In other embodiments, the effective dose is less than about 350 ng/cm 2 .
  • the effective dose is about 35 ng/cm 2 , or about 50 ng/cm 2 , or about 75 ng/cm 2 , or about 100 ng/cm 2 , or about 150 ng/cm 2 , or about 200 ng/cm 2 , or about 250 ng/cm 2 , or about 300 ng/cm 2 , or about 350 ng/cm 2 .
  • the effective dose of the nucleic acid drug is about 35 ng/cm 2 to about 7000 ng/cm 2 , or about 50 ng/cm 2 to about 5000 ng/cm 2 , or about 100 ng/cm 2 to about 3000 ng/cm 2 , or about 500 ng/cm 2 to about 2000 ng/cm 2 , or about 750 ng/cm 2 to about 1500 ng/cm 2 , or about 800 ng/cm 2 to about 1200 ng/cm 2 , or about 900 ng/cm 2 to about 1100 ng/cm 2 .
  • the effective dose of the nucleic acid drug is about 1 picomole/cm 2 , or about 2 picomoles/cm 2 , or about 3 picomoles/cm 2 , or about 4 picomoles/cm 2 , or about 5 picomoles/cm 2 , or about 6 picomoles/cm 2 , or about 7 picomoles/cm 2 , or about 8 picomoles/cm 2 , or about 9 picomoles/cm 2 , or about 10 picomoles/cm 2 , or about 12 picomoles/cm 2 , or about 14 picomoles/cm 2 , or about 16 picomoles/cm 2 , or about 18 picomoles/cm 2 , or about 20 picomoles/cm 2 .
  • the effective dose is less than about 1 picomole/cm 2 .
  • the effective dose is about 0.1 picomoles/cm 2 , or about 0.2 picomoles/cm 2 , or about 0.3 picomoles/cm 2 , or about 0.4 picomoles/cm 2 , or about 0.5 picomoles/cm 2 , or about 0.6 picomoles/cm 2 , or about 0.7 picomoles/cm 2 , or about 0.8 picomoles/cm 2 , or about 0.9 picomoles/cm 2 , or about 1 picomole/cm 2 .
  • the effective dose of the nucleic acid drug is about 0.1 picomoles/cm 2 to about 20 picomoles/cm 2 , or about 0.2 picomoles/cm 2 to about 15 picomoles/cm 2 , or about 0.5 picomoles/cm 2 to about 10 picomoles/cm 2 , or about 0.8 picomoles/cm 2 to about 8 picomoles/cm 2 , or about 1 picomole/cm 2 to about 5 picomoles/cm 2 , or about 2 picomoles/cm 2 to about 4 picomoles/cm 2 .
  • the nucleic acid drug, including synthetic RNA is administered in a pharmaceutically acceptable formulation.
  • the nucleic acid drug, including synthetic RNA is formulated for one or more of injection and topical administration.
  • the nucleic acid drug, including synthetic RNA may be formulated for injection to a tissue of interest, e.g. a disease site (by way of non-limiting example, a tumor).
  • injection includes delivery via a patch.
  • the delivery is mediated by electrical stimulation.
  • the nucleic acid drug is formulated for administration to one or more of the epidermis (optionally selected from the stratum corneum, stratum lucidum, stratum granulosum, stratum spinosum, and stratum germinativum), basement membrane, dermis (optionally selected from the papillary region and the reticular region), subcutis, conjunctiva cornea, sclera, iris, lens, corneal limbus, optic nerve, choroid, ciliary body, anterior segment, anterior chamber, and retina.
  • the epidermis optionally selected from the stratum corneum, stratum lucidum, stratum granulosum, stratum spinosum, and stratum germinativum
  • basement membrane optionally selected from the papillary region and the reticular region
  • dermis optionally selected from the papillary region and the reticular region
  • subcutis conjunctiva cornea, sclera, iris, lens, corneal limbus, optic nerve, choroid
  • the nucleic acid drug, including synthetic RNA is formulated for one or more of subcutaneous injection, intradermal injection, subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection, and topical administration.
  • the nucleic acid drug, including synthetic RNA is formulated for intradermal (ID) injection to one or more of the dermis or epidermis.
  • the nucleic acid drug, including synthetic RNA is administered in a manner such that it effects one or more of keratinocytes and fibroblasts (e.g. causes these cells to express one or more therapeutic proteins).
  • the present invention provides various formulations as described herein.
  • the formulations described herein find use in the various delivery and/or treatment methods of the present invention.
  • formulations can comprise a vesicle, for instance, a liposome (see Langer, 1990 , Science 249:1527-1533; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer , Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989).
  • the formulation comprises an aqueous suspension of liposomes.
  • Illustrative liposome components are set forth in Table 1, and are given by way of example, and not by way of limitation.
  • one or more, or two or more, or three or more, or four or more, or five or more of the lipids of Table 1 are combined in a formulation.
  • the liposomes include LIPOFECTAMINE 3000.
  • the liposomes include one or more lipids described in U.S. Pat. No. 4,897,355 or 7,479,573 or in International Patent Publication No. WO/2015/089487, or in Feigner, P. L. et al. (1987) Proc. Natl. Acad. Sci. USA 84:7413-7417, the entire contents of each is incorporated by reference in their entireties).
  • the liposome comprises N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA). In some embodiments, the liposome comprises dioleoylphosphatidylethanolamine (DOPE).
  • DOPE dioleoylphosphatidylethanolamine
  • the liposomes include one or more polyethylene glycol (PEG) chains, optionally selected from PEG200, PEG300, PEG400, PEG600, PEG800, PEG1000, PEG1500, PEG2000, PEG3000, and PEG4000.
  • PEG polyethylene glycol
  • the PEG is PEG2000.
  • the liposomes include 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) or a derivative thereof.
  • DSPE 1,2-distearoyl-sn-glycero-3-phosphoethanolamine
  • the formulation comprises one or more of N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-DSPE), fully hydrogenated phosphatidylcholine, cholesterol, LIPOFECTAMINE 3000, a cationic lipid, a polycationic lipid, and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[folate(polyethylene glycol)-5000] (FA-MPEG5000-DSPE).
  • MPEG2000-DSPE N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine
  • FA-MPEG5000-DSPE 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[folate(polyethylene glycol)-5000]
  • the formulation comprises about 3.2 mg/mL N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-DSPE), about 9.6 mg/mL fully hydrogenated phosphatidylcholine, about 3.2 mg/mL cholesterol, about 2 mg/mL ammonium sulfate, and histidine as a buffer, with about 0.27 mg/mL 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[folate(polyethylene glycol)-5000](FA-MPEG5000-DSPE) added to the lipid mixture.
  • MPEG2000-DSPE N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine
  • the nucleic acids are complexed by combining 1 ⁇ L of LIPOFECTAMINE 3000 per about 1 ⁇ g of nucleic acid and incubating at room temperature for at least about 5 minutes.
  • the LIPOFECTAMINE 3000 is a solution comprising a lipid at a concentration of about 1 mg/mL.
  • nucleic acids are encapsulated by combining about 10 ⁇ g of the liposome formulation per about 1 ⁇ g of nucleic acid and incubating at room temperature for about 5 minutes.
  • the formulation comprises one or more nanoparticles.
  • the nanoparticle is a polymeric nanoparticle.
  • the formulation comprises one or more of a diblock copolymer, a triblock copolymer, a tetrablock copolymer, and a multiblock copolymer.
  • the formulation comprises one or more of polymeric nanoparticles comprising a polyethylene glycol (PEG)-modified polylactic acid (PLA) diblock copolymer (PLA-PEG) and PEG-polypropylene glycol-PEG-modified PLA-tetrablock copolymer (PLA-PEG-PPG-PEG).
  • the formulation comprises one or more lipids that are described in WO/2000/027795, the entire contents of which are hereby incorporated by reference.
  • the therapeutic comprises one or more ligands.
  • the therapeutic comprises at least one of: androgen, CD30 (TNFRSF8), a cell-penetrating peptide, CXCR, estrogen, epidermal growth factor, EGFR, HER2, folate, insulin, insulin-like growth factor-I, interleukin-13, integrin, progesterone, stromal-derived-factor-1, thrombin, vitamin D, and transferrin or a biologically active fragment or variant thereof.
  • compositions of the present invention may include classic pharmaceutical preparations. Administration of these compositions according to the present invention may be via any common route so long as the target tissue is available via that route. This includes oral, nasal, or buccal. Alternatively, administration may be by intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection, or by direct injection into diseased, e.g. cancer, tissue.
  • the agents disclosed herein may also be administered by catheter systems. Such compositions would normally be administered as pharmaceutically acceptable compositions as described herein.
  • Administration of the compositions described herein may be, for example, by injection, topical administration, ophthalmic administration and intranasal administration.
  • the injection in some embodiments, may be linked to an electrical force (e.g. electroporation, including with devices that find use in electrochemotherapy (e.g. CLINIPORATOR, IGEA Srl, Carpi [MO], Italy)).
  • the topical administration may be, but is not limited to, a cream, lotion, ointment, gel, spray, solution and the like.
  • the topical administration may further include a penetration enhancer such as, but not limited to, surfactants, fatty acids, bile salts, chelating agents, non-chelating non-surfactants, polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether, fatty acids and/or salts in combination with bile acids and/or salts, sodium salt in combination with lauric acid, capric acid and UDCA, and the like.
  • the topical administration may also include a fragrance, a colorant, a sunscreen, an antibacterial and/or a moisturizer.
  • compositions described herein may be administered to at least one site such as, but not limited to, forehead, scalp, hair follicles, hair, upper eyelids, lower eyelids, eyebrows, eyelashes, infraorbital area, periorbital areas, temple, nose, nose bridge, cheeks, tongue, nasolabial folds, lips, periobicular areas, jaw line, ears, neck, breast, forearm, upper arm, palm, hand, finger, nails, back, abdomen, sides, buttocks, thigh, calf, feet, toes and the like.
  • site such as, but not limited to, forehead, scalp, hair follicles, hair, upper eyelids, lower eyelids, eyebrows, eyelashes, infraorbital area, periorbital areas, temple, nose, nose bridge, cheeks, tongue, nasolabial folds, lips, periobicular areas, jaw line, ears, neck, breast, forearm, upper arm, palm, hand, finger,
  • Routes of administration include, for example: intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, or topically, particularly to the ears, nose, eyes, or skin.
  • the administering is effected orally or by parenteral injection.
  • solutions may be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective, as described herein.
  • the formulations may easily be administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
  • aqueous solution for example, the solution generally is suitably buffered and the liquid diluent first rendered isotonic with, for example, sufficient saline or glucose.
  • aqueous solutions may be used, for example, for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media are employed as is known to those of skill in the art, particularly in light of the present disclosure.
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose is administered to a surface area of about 4 mm 2 to about 150 mm 2 (e.g. about, or no more than about, 4 mm 2 , or about 5 mm 2 , or about 6 mm 2 , or about 7 mm 2 , or about 8 mm 2 , or about 10 mm 2 , or about 20 mm 2 , or about 50 mm 2 , or about 100 mm 2 , or about 150 mm 2 ).
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose administered to a surface area of no more than about 4 mm 2 , or about 5 mm 2 , or about 6 mm 2 , or about 7 mm 2 , or about 8 mm 2 , or about 10 mm 2 , or about 20 mm 2 , or about 50 mm 2 , or about 100 mm 2 , or about 150 mm 2 .
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose administered to a surface area of about 4 mm 2 , or about 5 mm 2 , or about 6 mm 2 , or about 7 mm 2 , or about 8 mm 2 , or about 10 mm 2 , or about 20 mm 2 , or about 50 mm 2 , or about 100 mm 2 , or about 150 mm 2 .
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose (weight RNA/surface area of injection) is about 35 ng/cm 2 to about 7000 ng/cm 2 .
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose (weight RNA/surface area of injection) is no more than about 35 ng/cm 2 , or about 50 ng/cm 2 , or about 75 ng/cm 2 , or about 100 ng/cm 2 , or about 125 ng/cm 2 , or about 150 ng/cm 2 , or about 175 ng/cm 2 , or about 200 ng/cm 2 , or about 225 ng/cm 2 , or about 250 ng/cm 2 , or about 500 ng/cm 2 , or about 1000 ng/cm 2 , or about 2000 ng/cm 2 , or about 5000 ng/cm 2 , or about 7000 ng/cm
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose (weight RNA/surface area of injection) is about 35 ng/cm 2 , or about 50 ng/cm 2 , or about 75 ng/cm 2 , or about 100 ng/cm 2 , or about 125 ng/cm 2 , or about 150 ng/cm 2 , or about 175 ng/cm 2 , or about 200 ng/cm 2 , or about 225 ng/cm 2 , or about 250 ng/cm 2 , or about 500 ng/cm 2 , or about 1000 ng/cm 2 , or about 2000 ng/cm 2 , or about 5000 ng/cm 2 , or about 7000 ng/cm 2 .
  • compositions may additionally comprise delivery reagents (a.k.a. “transfection reagents”, a.k.a. “vehicles”, a.k.a. “delivery vehicles”) and/or excipients.
  • delivery reagents a.k.a. “transfection reagents”, a.k.a. “vehicles”, a.k.a. “delivery vehicles”
  • excipients a.k.a. “delivery vehicles”
  • Pharmaceutically acceptable delivery reagents, excipients, and methods of preparation and use thereof, including methods for preparing and administering pharmaceutical preparations to patients (a.k.a. “subjects”) are well known in the art, and are set forth in numerous publications, including, for example, in US Patent Appl. Pub. No. US 2008/0213377, the entirety of which is incorporated herein by reference.
  • compositions can be in the form of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salts include those listed in, for example, J. Pharma. Sci. 66, 2-19 (1977) and The Handbook of Pharmaceutical Salts; Properties, Selection, and Use . P. H. Stahl and C. G. Wermuth (eds.), Verlag, Zurich (Switzerland) 2002, which are hereby incorporated by reference in their entirety.
  • Non-limiting examples of pharmaceutically acceptable salts include: sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, pamoate, phenylacetate, trifluoroacetate, acrylate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate,
  • the present pharmaceutical compositions can comprises excipients, including liquids such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • the pharmaceutical excipients can be, for example, saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like.
  • auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used.
  • the pharmaceutically acceptable excipients are sterile when administered to a subject.
  • Suitable pharmaceutical excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Any agent described herein, if desired, can also comprise minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Dosage forms suitable for parenteral administration include, for example, solutions, suspensions, dispersions, emulsions, and the like. They may also be manufactured in the form of sterile solid compositions (e.g. lyophilized composition), which can be dissolved or suspended in sterile injectable medium immediately before use. They may contain, for example, suspending or dispersing agents known in the art.
  • the formulations described herein may comprise albumin and a nucleic acid molecule.
  • the invention relates to a cosmetic composition.
  • the cosmetic composition comprises albumin.
  • the albumin is treated with an ion-exchange resin or charcoal.
  • the cosmetic composition comprises a nucleic acid molecule.
  • the nucleic acid molecule encodes a member of the group: elastin, collagen, tyrosinase, melanocortin 1 receptor, keratin, filaggren, an antibody, and hyaluronan synthase or a biologically active fragment, variant, analogue or family member thereof.
  • the present invention provides treatment regimens.
  • the inventors have discovered that the doses and administration described herein can produce a substantial protein expression effect quickly (e.g. in about 6, or about 12, or about 24, or about 36, or about 48 hours). Further, these effects can be sustained for about 7 days, or longer.
  • the present methods provide for administration of a nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, about weekly to about once every 20 weeks.
  • the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides is administered about weekly, for at least 2 weeks (e.g. 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10 weeks). In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered about every other week for at least one month (e.g. 1, or 2, or 3, or 4, or 5, or 6, or 12 months). In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered monthly or about every other month.
  • the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered is administered for at least two months, or at least 4 months, or at least 6 months, or at least 9 months, or at least one year.
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, is administered about weekly, or about once every 2 weeks, or about once every 3 weeks, or about once every 4 weeks, or about once every 5 weeks, or about once every 6 weeks, or about once every 7 weeks, or about once every 8 weeks, or about once every 9 weeks, or about once every 10 weeks, or about once every 11 weeks, or about once every 12 weeks, or about once every 13 weeks, or about once every 14 weeks, or about once every 15 weeks, or about once every 20 weeks, or about once every 24 weeks.
  • the nucleic acid drug including RNA comprising one or more non-canonical nucleotides, is administered no more than about weekly, or about once every 2 weeks, or about once every 3 weeks, or about once every 4 weeks, or about once every 5 weeks, or about once every 6 weeks, or about once every 7 weeks, or about once every 8 weeks, or about once every 9 weeks, or about once every 10 weeks, or about once every 11 weeks, or about once every 12 weeks, or about once every 13 weeks, or about once every 14 weeks, or about once every 15 weeks, or about once every 20 weeks, or about 24 weeks.
  • Certain proteins have long half-lives, and can persist in tissues for several hours, days, weeks, months or years. It has now been discovered that certain methods of treating a patient can result in accumulation of one or more proteins, including, for example, one or more beneficial proteins. Certain embodiments are therefore directed to a method for treating a patient comprising delivering to a patient in a series of doses a nucleic acid encoding one or more proteins.
  • the nucleic acid comprises RNA comprising one or more non-canonical nucleotides.
  • a first dose is given at a first time-point.
  • a second dose is given at a second time-point.
  • the amount of at least one of the one or more proteins in the patient at the second time-point is greater than the amount of said protein at the first time-point.
  • the method results in the accumulation of said protein in the patient.
  • the present invention relates to nucleic acid drugs, which, in various embodiments are RNA comprising one or more non-canonical nucleotides.
  • Certain non-canonical nucleotides when incorporated into RNA molecules, can reduce the toxicity of the RNA molecules, in part, without wishing to be bound by theory, by interfering with binding of proteins that detect exogenous nucleic acids, for example, protein kinase R, Rig-1 and the oligoadenylate synthetase family of proteins.
  • Non-canonical nucleotides that have been reported to reduce the toxicity of RNA molecules when incorporated therein include: pseudouridine, 5-methyluridine, 2-thiouridine, 5-methylcytidine, N6-methyladenosine, and certain combinations thereof.
  • pseudouridine 5-methyluridine
  • 2-thiouridine 5-methylcytidine
  • N6-methyladenosine N6-methyladenosine
  • RNA molecules containing these nucleotides can reduce the efficiency with which RNA molecules can be translated into protein, limiting the utility of RNA molecules containing these nucleotides in applications that require protein expression.
  • pseudouridine can be completely substituted for uridine in RNA molecules without reducing the efficiency with which the synthetic RNA molecules can be translated into protein, in certain situations, for example, when performing frequent, repeated transfections, synthetic RNA molecules containing only adenosine, guanosine, cytidine, and pseudouridine can exhibit excessive toxicity.
  • RNA molecules containing one or more non-canonical nucleotides that include one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine can be less toxic than synthetic RNA molecules containing only canonical nucleotides, due in part to the ability of substitutions at these positions to interfere with recognition of synthetic RNA molecules by proteins that detect exogenous nucleic acids, and furthermore, that substitutions at these positions can have minimal impact on the efficiency with which the synthetic RNA molecules can be translated into protein, due in part to the lack of interference of substitutions at these positions with base-pairing and base-stacking interactions.
  • non-canonical nucleotides that include one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine include, but are not limited to: 2-thiouridine, 5-azauridine, pseudouridine, 4-thiouridine, 5-methyluridine, 5-methylpseudouridine, 5-aminouridine, 5-aminopseudouridine, 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methoxyuridine, 5-methoxypseudouridine, 5-hydroxymethyluridine, 5-hydroxymethylpseudouridine, 5-carboxyuridine, 5-carboxypseudouridine, 5-formyluridine, 5-formylpseudouridine, 5-methyl-5-azauridine, 5-amino-5-azauridine, 5-hydroxy-5-azauridine, 5-methylpseudouridine, 5-aminopse
  • the invention relates to one or more non-canonical nucleotides selected from 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, 5-hydroxyuridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine.
  • At least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of the non-canonical nucleotides are one or more of 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine.
  • At least about 50%, or at least about 55%%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of cytidine residues are non-canonical nucleotides selected from 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine.
  • At least about 20%, or about 30%, or about 40%, or about 50%, or at least about 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of uridine residues are non-canonical nucleotides selected from 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine.
  • At least about 10% (e.g. 10%, or about 20%, or about 30%, or about 40%, or about 50%) of guanosine residues are non-canonical nucleotides, and the non-canonical nucleotide is optionally 7-deazaguanosine.
  • the RNA contains no more than about 50% 7-deazaguanosine in place of guanosine residues.
  • the RNA does not contain non-canonical nucleotides in place of adenosine residues.
  • 5-methylpseudouridine can be referred to as “3-methylpseudouridine” or “N3-methylpseudouridine” or “1-methylpseudouridine” or “N1-methylpseudouridine”.
  • Nucleotides that contain the prefix “amino” can refer to any nucleotide that contains a nitrogen atom bound to the atom at the stated position of the nucleotide, for example, 5-aminocytidine can refer to 5-aminocytidine, 5-methylaminocytidine, and 5-nitrocytidine.
  • nucleotides that contain the prefix “methyl” can refer to any nucleotide that contains a carbon atom bound to the atom at the stated position of the nucleotide
  • 5-methylcytidine can refer to 5-methylcytidine, 5-ethylcytidine, and 5-hydroxymethylcytidine
  • nucleotides that contain the prefix “thio” can refer to any nucleotide that contains a sulfur atom bound to the atom at the given position of the nucleotide
  • nucleotides that contain the prefix “hydroxy” can refer to any nucleotide that contains an oxygen atom bound to the atom at the given position of the nucleotide
  • 5-hydroxyuridine can refer to 5-hydroxyuridine and uridine with a methyl group bound to an oxygen atom, wherein the oxygen atom is bound to the atom at the 5C position of the uridine.
  • RNA comprising one or more non-canonical nucleotides, wherein the RNA molecule contains one or more nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine.
  • the therapeutic contains one or more RNA molecules comprising one or more non-canonical nucleotides, and wherein the one or more RNA molecules comprising one or more non-canonical nucleotides contains one or more nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine.
  • the therapeutic comprises a transfection reagent.
  • the transfection reagent comprises a cationic lipid, liposome or micelle.
  • the liposome or micelle comprises folate and the therapeutic composition has anti-cancer activity.
  • the one or more nucleotides includes at least one of pseudouridine, 2-thiouridine, 4-thiouridine, 5-azauridine, 5-hydroxyuridine, 5-methyluridine, 5-aminouridine, 2-thiopseudouridine, 4-thiopseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-aminopseudouridine, pseudoisocytidine, N4-methylcytidine, 2-thiocytidine, 5-azacytidine, 5-hydroxycytidine, 5-aminocytidine, 5-methylcytidine, N4-methylpseudoisocytidine, 2-thiopseudoisocytidine, 5-hydroxypseudoisocytidine, 5-aminopseudoisocytidine, 5-methylpseudoisocytidine, 7-deaza
  • the one or more nucleotides includes at least one of pseudouridine, 2-thiouridine, 4-thiouridine, 5-azauridine, 5-hydroxyuridine, 5-methyluridine, 5-aminouridine, 2-thiopseudouridine, 4-thiopseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, and 5-aminopseudouridine and at least one of pseudoisocytidine, N4-methylcytidine, 2-thiocytidine, 5-azacytidine, 5-hydroxycytidine, 5-aminocytidine, 5-methylcytidine, N4-methylpseudoisocytidine, 2-thiopseudoisocytidine, 5-hydroxypseudoisocytidine, 5-aminopseudoisocytidine, and 5-methylpseudoisocytidine.
  • the one or more nucleotides include at least one of pseudouridine, 2-thiouridine, 4-thiouridine, 5-azauridine, 5-hydroxyuridine, 5-methyluridine, 5-aminouridine, 2-thiopseudouridine, 4-thiopseudouridine, 5-hydroxypseudouridine, and 5-methylpseudouridine, 5-aminopseudouridine and at least one of pseudoisocytidine, N4-methylcytidine, 2-thiocytidine, 5-azacytidine, 5-hydroxycytidine, 5-aminocytidine, 5-methylcytidine, N4-methylpseudoisocytidine, 2-thiopseudoisocytidine, 5-hydroxypseudoisocytidine, 5-aminopseudoisocytidine, and 5-methylpseudoisocytidine and at least one of 7-deazaguanosine, 6-thioguanosine, 6-thiouridine, 5-
  • the one or more nucleotides includes: 5-methylcytidine and 7-deazaguanosine.
  • the one or more nucleotides also includes pseudouridine or 4-thiouridine or 5-methyluridine or 5-aminouridine or 4-thiopseudouridine or 5-methylpseudouridine or 5-aminopseudouridine.
  • the one or more nucleotides also includes 7-deazaadenosine.
  • the one or more nucleotides includes: pseudoisocytidine and 7-deazaguanosine and 4-thiouridine.
  • the one or more nucleotides includes: pseudoisocytidine or 7-deazaguanosine and pseudouridine. In still another embodiment, the one or more nucleotides includes: 5-methyluridine and 5-methylcytidine and 7-deazaguanosine. In a further embodiment, the one or more nucleotides includes: pseudouridine or 5-methylpseudouridine and 5-methylcytidine and 7-deazaguanosine. In another embodiment, the one or more nucleotides includes: pseudoisocytidine and 7-deazaguanosine and pseudouridine. In one embodiment, the RNA comprising one or more non-canonical nucleotides is present in vivo.
  • Certain non-canonical nucleotides can be incorporated more efficiently than other non-canonical nucleotides into RNA molecules by RNA polymerases that are commonly used for in vitro transcription, due in part to the tendency of these certain non-canonical nucleotides to participate in standard base-pairing interactions and base-stacking interactions, and to interact with the RNA polymerase in a manner similar to that in which the corresponding canonical nucleotide interacts with the RNA polymerase.
  • certain nucleotide mixtures containing one or more non-canonical nucleotides can be beneficial in part because in vitro-transcription reactions containing these nucleotide mixtures can yield a large quantity of RNA.
  • Certain embodiments are therefore directed to a nucleotide mixture containing one or more nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine.
  • Nucleotide mixtures include, but are not limited to (numbers preceding each nucleotide indicate an exemplary fraction of the non-canonical nucleotide triphosphate in an in vitro-transcription reaction, for example, 0.2 pseudoisocytidine refers to a reaction containing adenosine-5′-triphosphate, guanosine-5′-triphosphate, uridine-5′-triphosphate, cytidine-5′-triphosphate, and pseudoisocytidine-5′-triphosphate, wherein pseudoisocytidine-5′-triphosphate is present in the reaction at an amount approximately equal to 0.2 times the total amount of pseudoisocytidine-5′-triphosphate+cytidine-5′-triphosphate that is present in the reaction, with amounts measured either on a molar or mass basis, and wherein more than one number preceding a nucleoside indicates a range of exemplary fractions): 1.0 pseudouridine, 0.1-0.8 2-thi
  • the RNA comprising one or more non-canonical nucleotides composition or synthetic polynucleotide composition contains substantially or entirely the canonical nucleotide at positions having adenine or “A” in the genetic code.
  • the term “substantially” in this context refers to at least 90%.
  • the RNA composition or synthetic polynucleotide composition may further contain (e.g., consist of) 7-deazaguanosine at positions with “G” in the genetic code as well as the corresponding canonical nucleotide “G”, and the canonical and non-canonical nucleotide at positions with G may be in the range of 5:1 to 1:5, or in some embodiments in the range of 2:1 to 1:2.
  • the RNA composition or synthetic polynucleotide composition may further contain (e.g., consist of) one or more (e.g., two, three or four) of 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine at positions with “C” in the genetic code as well as the canonical nucleotide “C”, and the canonical and non-canonical nucleotide at positions with C may be in the range of 5:1 to 1:5, or in some embodiments in the range of 2:1 to 1:2.
  • the level of non-canonical nucleotide at positions of “C” are as described in the preceding paragraph.
  • the RNA composition or synthetic polynucleotide composition may further contain (e.g., consist of) one or more (e.g., two, three, or four) of 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine at positions with “U” in the genetic code as well as the canonical nucleotide “U”, and the canonical and non-canonical nucleotide at positions with “U” may be in the range of 5:1 to 1:5, or in some embodiments in
  • nucleotide mixture contains more than one of the non-canonical nucleotides listed above, for example, the nucleotide mixture contains both pseudoisocytidine and 7-deazaguanosine or the nucleotide mixture contains both N4-methylcytidine and 7-deazaguanosine, etc.
  • the nucleotide mixture contains more than one of the non-canonical nucleotides listed above, and each of the non-canonical nucleotides is present in the mixture at the fraction listed above, for example, the nucleotide mixture contains 0.1-0.8 pseudoisocytidine and 0.2-1.0 7-deazaguanosine or the nucleotide mixture contains 0.2-1.0 N4-methylcytidine and 0.2-1.0 7-deazaguanosine, etc.
  • nucleotide fractions other than those given above may be used.
  • the exemplary fractions and ranges of fractions listed above relate to nucleotide-triphosphate solutions of typical purity (greater than 90% purity). Larger fractions of these and other nucleotides can be used by using nucleotide-triphosphate solutions of greater purity, for example, greater than about 95% purity or greater than about 98% purity or greater than about 99% purity or greater than about 99.5% purity, which can be achieved, for example, by purifying the nucleotide triphosphate solution using existing chemical-purification technologies such as high-pressure liquid chromatography (HPLC) or by other means.
  • nucleotides with multiple isomers are purified to enrich the desired isomer.
  • RNA molecules that contains one or more non-canonical nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine.
  • Still other embodiments are directed to a method for transfecting, reprogramming, and/or gene-editing a cell in vivo by contacting the cell with a RNA molecule that contains one or more non-canonical nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine.
  • the RNA molecule is produced by in vitro transcription.
  • the RNA molecule encodes one or more reprogramming factors.
  • the one or more reprogramming factors includes Oct4 protein.
  • the cell is also contacted with a RNA molecule that encodes Sox2 protein. In yet another embodiment, the cell is also contacted with a RNA molecule that encodes Klf4 protein. In yet another embodiment, the cell is also contacted with a RNA molecule that encodes c-Myc protein. In yet another embodiment, the cell is also contacted with a RNA molecule that encodes Lin28 protein.
  • Enzymes such as T7 RNA polymerase may preferentially incorporate canonical nucleotides in an in vitro-transcription reaction containing both canonical and non-canonical nucleotides.
  • an in vitro-transcription reaction containing a certain fraction of a non-canonical nucleotide may yield RNA containing a different, often lower, fraction of the non-canonical nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction.
  • references to nucleotide incorporation fractions therefore can refer both to RNA molecules containing the stated fraction of the nucleotide, and to RNA molecules synthesized in a reaction containing the stated fraction of the nucleotide (or nucleotide derivative, for example, nucleotide-triphosphate), even though such a reaction may yield RNA containing a different fraction of the nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction.
  • references to nucleotide incorporation fractions therefore can refer both to RNA molecules containing the stated fraction of the nucleotide, and to RNA molecules encoding the same protein as a different RNA molecule, wherein the different RNA molecule contains the stated fraction of the nucleotide.
  • kits containing one or more materials needed to practice the present invention.
  • the kit contains one or more synthetic RNA molecules.
  • the kit contains one or more synthetic RNA molecules that encode one or more reprogramming factors and/or gene-editing proteins.
  • the one or more synthetic RNA molecules contain one or more non-canonical nucleotides that include one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine.
  • the kit contains one or more of: a transfection medium, a transfection reagent, a complexation medium, and a matrix solution.
  • the matrix solution contains fibronectin and/or vitronectin or recombinant fibronectin and/or recombinant vitronectin.
  • one or more of the components of the kit are present as a plurality of aliquots.
  • the kit contains aliquots of nucleic acid transfection-reagent complexes.
  • the kit contains aliquots of nucleic acid transfection-reagent complexes that are provided in a solid form, for example, as frozen or freeze-dried pellets.
  • the kit contains aliquots of medium, wherein each aliquot contains transfection reagent-nucleic acid complexes that are stabilized either by chemical treatment or by freezing.
  • Transfection in general, and reprogramming, in particular, can be difficult and time-consuming techniques that can be repetitive and prone to error. However, these techniques are often performed manually due to the lack of automated transfection equipment. Certain embodiments are therefore directed to a system that can transfect, reprogram, and/or gene-edit cells in vivo in an automated or semi-automated manner.
  • non-canonical nucleotide members of the 5-methylcytidine de-methylation pathway when incorporated into synthetic RNA, can increase the efficiency with which the synthetic RNA can be translated into protein in vivo, and can decrease the toxicity of the synthetic RNA in vivo.
  • These non-canonical nucleotides include, for example: 5-methylcytidine, 5-hydroxymethylcytidine, 5-formylcytidine, and 5-carboxycytidine (a.k.a. “cytidine-5-carboxylic acid”). Certain embodiments are therefore directed to a nucleic acid.
  • the nucleic acid is present in vivo.
  • the nucleic acid is a synthetic RNA molecule.
  • the nucleic acid comprises one or more non-canonical nucleotides.
  • the nucleic acid comprises one or more non-canonical nucleotide members of the 5-methylcytidine de-methylation pathway.
  • the nucleic acid comprises at least one of: 5-methylcytidine, 5-hydroxymethylcytidine, 5-formylcytidine, and 5-carboxycytidine or a derivative thereof.
  • the nucleic acid comprises at least one of: pseudouridine, 5-methylpseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, N4-methylcytidine, N4-acetylcytidine, and 7-deazaguanosine or a derivative thereof.
  • Certain embodiments are directed to a protein. Other embodiments are directed to a nucleic acid that encodes a protein.
  • the protein is a protein of interest.
  • the protein is selected from: a reprogramming protein and a gene-editing protein.
  • the nucleic acid is a plasmid.
  • the nucleic acid is present in a virus or viral vector.
  • the virus or viral vector is replication incompetent.
  • the virus or viral vector is replication competent.
  • the virus or viral vector includes at least one of: an adenovirus, a retrovirus, a lentivirus, a herpes virus, an adeno-associated virus or a natural or engineered variant thereof, and an engineered virus.
  • non-canonical nucleotides can be particularly effective at increasing the efficiency with which synthetic RNA can be translated into protein in vivo, and decreasing the toxicity of synthetic RNA in vivo, for example, the combinations: 5-methyluridine and 5-methylcytidine, 5-hydroxyuridine and 5-methylcytidine, 5-hydroxyuridine and 5-hydroxymethylcytidine, 5-methyluridine and 7-deazaguanosine, 5-methylcytidine and 7-deazaguanosine, 5-methyluridine, 5-methylcytidine, and 7-deazaguanosine, and 5-methyluridine, 5-hydroxymethylcytidine, and 7-deazaguanosine.
  • Certain embodiments are therefore directed to a nucleic acid comprising at least two of: 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, and 7-deazaguanosine or one or more derivatives thereof.
  • Other embodiments are directed to a nucleic acid comprising at least three of: 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, and 7-deazaguanosine or one or more derivatives thereof.
  • Other embodiments are directed to a nucleic acid comprising all of: 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, and 7-deazaguanosine or one or more derivatives thereof.
  • the nucleic acid comprises one or more 5-methyluridine residues, one or more 5-methylcytidine residues, and one or more 7-deazaguanosine residues or one or more 5-methyluridine residues, one or more 5-hydroxymethylcytidine residues, and one or more 7-deazaguanosine residues.
  • RNA molecules containing certain fractions of certain non-canonical nucleotides and combinations thereof can exhibit particularly high translation efficiency and low toxicity in vivo.
  • Certain embodiments are therefore directed to a nucleic acid comprising at least one of: one or more uridine residues, one or more cytidine residues, and one or more guanosine residues, and comprising one or more non-canonical nucleotides.
  • a nucleic acid comprising at least one of: one or more uridine residues, one or more cytidine residues, and one or more guanosine residues, and comprising one or more non-canonical nucleotides.
  • between about 20% and about 80% of the uridine residues are 5-methyluridine residues.
  • between about 30% and about 50% of the uridine residues are 5-methyluridine residues.
  • about 40% of the uridine residues are 5-methyluridine residues.
  • between about 60% and about 80% of the cytidine residues are 5-methylcytidine residues. In another embodiment, between about 80% and about 100% of the cytidine residues are 5-methylcytidine residues. In a further embodiment, about 100% of the cytidine residues are 5-methylcytidine residues. In a still further embodiment, between about 20% and about 100% of the cytidine residues are 5-hydroxymethylcytidine residues. In one embodiment, between about 20% and about 80% of the guanosine residues are 7-deazaguanosine residues. In another embodiment, between about 40% and about 60% of the guanosine residues are 7-deazaguanosine residues.
  • guanosine residues are 7-deazaguanosine residues. In one embodiment, between about 20% and about 80% or between about 30% and about 60% or about 40% of the cytidine residues are N4-methylcytidine and/or N4-acetylcytidine residues. In another embodiment, each cytidine residue is a 5-methylcytidine residue. In a further embodiment, about 100% of the cytidine residues are 5-methylcytidine residues and/or 5-hydroxymethylcytidine residues and/or N4-methylcytidine residues and/or N4-acetylcytidine residues and/or one or more derivatives thereof.
  • about 40% of the uridine residues are 5-methyluridine residues, between about 20% and about 100% of the cytidine residues are N4-methylcytidine and/or N4-acetylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues.
  • about 40% of the uridine residues are 5-methyluridine residues and about 100% of the cytidine residues are 5-methylcytidine residues.
  • about 40% of the uridine residues are 5-methyluridine residues and about 50% of the guanosine residues are 7-deazaguanosine residues.
  • cytidine residues are 5-methylcytidine residues and about 50% of the guanosine residues are 7-deazaguanosine residues.
  • about 100% of the uridine residues are 5-hydroxyuridine residues.
  • about 40% of the uridine residues are 5-methyluridine residues, about 100% of the cytidine residues are 5-methylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues.
  • each uridine residue in the synthetic RNA molecule is a pseudouridine residue or a 5-methylpseudouridine residue.
  • uridine residues are pseudouridine residues and/or 5-methylpseudouridine residues. In a further embodiment, about 100% of the uridine residues are pseudouridine residues and/or 5-methylpseudouridine residues, about 100% of the cytidine residues are 5-methylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues.
  • non-canonical nucleotides that can be used in place of or in combination with 5-methyluridine include, but are not limited to: pseudouridine, 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methoxyuridine, 5-methoxypseudouridine, 5-carboxyuridine, 5-carboxypseudouridine, 5-formyluridine, 5-formylpseudouridine, 5-hydroxymethyluridine, 5-hydroxymethylpseudouridine, and 5-methylpseudouridine (a.k.a. “1-methylpseudouridine”, a.k.a. “N1-methylpseudouridine”) or one or more derivatives thereof.
  • pseudouridine 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methoxyuridine, 5-methoxypseudouridine
  • 5-carboxyuridine 5-carboxypseudouridine
  • 5-formyluridine 5-formylps
  • non-canonical nucleotides that can be used in place of or in combination with 5-methylcytidine and/or 5-hydroxymethylcytidine include, but are not limited to: pseudoisocytidine, 5-methylpseudoisocytidine, 5-hydroxymethylcytidine, 5-formylcytidine, 5-carboxycytidine, 5-methoxycytidine, N4-methylcytidine, N4-acetylcytidine or one or more derivatives thereof.
  • the fractions of non-canonical nucleotides can be reduced. Reducing the fraction of non-canonical nucleotides can be beneficial, in part, because reducing the fraction of non-canonical nucleotides can reduce the cost of the nucleic acid. In certain situations, for example, when minimal immunogenicity of the nucleic acid is desired, the fractions of non-canonical nucleotides can be increased.
  • Enzymes such as T7 RNA polymerase may preferentially incorporate canonical nucleotides in an in vitro-transcription reaction containing both canonical and non-canonical nucleotides.
  • an in vitro-transcription reaction containing a certain fraction of a non-canonical nucleotide may yield RNA containing a different, often lower, fraction of the non-canonical nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction.
  • references to nucleotide incorporation fractions therefore can refer both to nucleic acids containing the stated fraction of the nucleotide, and to nucleic acids synthesized in a reaction containing the stated fraction of the nucleotide (or nucleotide derivative, for example, nucleotide-triphosphate), even though such a reaction may yield a nucleic acid containing a different fraction of the nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction.
  • nucleotide sequences can encode the same protein by utilizing alternative codons.
  • references to nucleotide incorporation fractions therefore can refer both to nucleic acids containing the stated fraction of the nucleotide, and to nucleic acids encoding the same protein as a different nucleic acid, wherein the different nucleic acid contains the stated fraction of the nucleotide.
  • nucleic acid comprising a 5′-cap structure selected from Cap 0, Cap 1, Cap 2, and Cap 3 or a derivative thereof.
  • the nucleic acid comprises one or more UTRs.
  • the one or more UTRs increase the stability of the nucleic acid.
  • the one or more UTRs comprise an alpha-globin or beta-globin 5′-UTR.
  • the one or more UTRs comprise an alpha-globin or beta-globin 3′-UTR.
  • the synthetic RNA molecule comprises an alpha-globin or beta-globin 5′-UTR and an alpha-globin or beta-globin 3′-UTR.
  • the 5′-UTR comprises a Kozak sequence that is substantially similar to the Kozak consensus sequence.
  • the nucleic acid comprises a 3′-poly(A) tail.
  • the 3′-poly(A) tail is between about 20 nt and about 250 nt or between about 120 nt and about 150 nt long.
  • the 3′-poly(A) tail is about 20 nt, or about 30 nt, or about 40 nt, or about 50 nt, or about 60 nt, or about 70 nt, or about 80 nt, or about 90 nt, or about 100 nt, or about 110 nt, or about 120 nt, or about 130 nt, or about 140 nt, or about 150 nt, or about 160 nt, or about 170 nt, or about 180 nt, or about 190 nt, or about 200 nt, or about 210 nt, or about 220 nt, or about 230 nt, or about 240 nt, or about 250 nt long.
  • Certain embodiments are directed to methods of making nucleic acid drugs, including RNA comprising one or more non-canonical nucleotides. Such methods yield substantially stable RNA.
  • the present methods and compositions find use in methods of treating, preventing or ameliorating a disease, disorder and/or condition.
  • the described methods of in vivo delivery including various effective doses, administration strategies and formulations are used in a method of treatment.
  • the present methods and compositions find use in methods of altering, modifying and/or changing a tissue (e.g. cosmetically).
  • the present methods and compositions include using a nucleic acid drug, including a synthetic RNA, in the diagnosing, treating, preventing or ameliorating of a disease, disorder and/or condition described herein.
  • the present methods and compositions include using a nucleic acid drug, including a synthetic RNA, in the altering, modifying and/or changing of a tissue (e.g. cosmetically).
  • a synthetic RNA as described herein is administered to a human at specific doses described herein and the synthetic RNA comprises a sequence, sometimes referred to as a target sequence that encodes a protein of interest, which may be a therapeutic protein.
  • Synthetic RNA comprising only canonical nucleotides can bind to pattern recognition receptors, can be recognized as a pathogen-associated molecular pattern, and can trigger a potent immune response in cells, which can result in translation block, the secretion of inflammatory cytokines, and cell death. It has now been discovered that synthetic RNA comprising certain non-canonical nucleotides can evade detection by the innate immune system, and can be translated at high efficiency into protein, including in humans.
  • synthetic RNA comprising at least one of the non-canonical nucleotides described herein, including, for example, a member of the group: 5-methylcytidine, 5-hydroxycytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-methoxyuridine, 5-formyluridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-methoxypseudouridine, and 5-formylpseudouridine can evade detection by the innate immune system, and can be translated at high efficiency into protein, including in humans.
  • Certain embodiments are therefore directed to a method for inducing a cell to express a protein of interest comprising contacting a cell with synthetic RNA.
  • Other embodiments are directed to a method for transfecting a cell with synthetic RNA comprising contacting a cell with a solution comprising one or more synthetic RNA molecules.
  • Still other embodiments are directed to a method for treating a patient comprising administering to the patient synthetic RNA.
  • the synthetic RNA comprises at least one of the non-canonical nucleotides described herein, including, for example, a member of the group: 5-methylcytidine, 5-hydroxycytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-methoxyuridine, 5-formyluridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-methoxypseudouridine, and 5-formylpseudouridine.
  • the synthetic RNA encodes a protein of interest.
  • Exemplary RNAs may contain combinations and levels of non-canonical and non-canonical nucleotides as described elsewhere herein, including with respect to the expression of any protein of interest described herein.
  • the method results in the expression of the protein of interest.
  • the method results in the expression of the protein of interest in the patient's skin.
  • inventions are directed to a method for delivering a nucleic acid to a cell in vivo. Still other embodiments are directed to a method for inducing a cell in vivo to express a protein of interest. Still other embodiments are directed to a method for treating a patient. In one embodiment, the method comprises disrupting the stratum corneum. In another embodiment, the method comprises contacting a cell with a nucleic acid. In yet another embodiment, the method results in the cell internalizing the nucleic acid. In a further embodiment, the method results in the cell expressing the protein of interest. In a still further embodiment, the method results in the expression of the protein of interest in the patient. In a still further embodiment, the method results in the amelioration of one or more of the patient's symptoms. In a still further embodiment, the patient is in need of the protein of interest. In a still further embodiment, the patient is deficient in the protein of interest.
  • Still other embodiments are directed to a method for treating a patient comprising delivering to a patient a composition.
  • the composition comprises albumin that is treated with an ion-exchange resin or charcoal.
  • the composition comprises one or more nucleic acid molecules.
  • at least one of the one or more nucleic acid molecules encodes a protein of interest.
  • the method results in the expression of the protein in the patient's skin.
  • the method results in the expression of a therapeutically or cosmetically effective amount of the protein of interest in the patient.
  • the method comprises administering a steroid.
  • the steroid is a member of the group: hydrocortisone and dexamethasone.
  • Some embodiments are directed to a therapeutic composition and/or methods of treatment comprising a nucleic acid molecule encoding one or more proteins, wherein at least one of the one or more proteins is an extracellular matrix protein. Still other embodiments are directed to a cosmetic composition comprising a nucleic acid molecule encoding one or more proteins, wherein at least one of the one or more proteins is an extracellular matrix protein.
  • Pigmentation disorders can cause severe symptoms in patients. It has now been discovered that pigmentation disorders can be treated by delivering to a patient a nucleic acid encoding tyrosinase. Certain embodiments are therefore directed to a method for treating a pigmentation disorder. Other embodiments are directed to a method for altering the pigmentation of a patient. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding tyrosinase. Other embodiments are directed to a cosmetic composition comprising a nucleic acid encoding tyrosinase. Still other embodiments are directed to a therapeutic composition comprising a nucleic acid encoding tyrosinase.
  • Still other embodiments are directed to a method for increasing the ultraviolet absorption of a patient's skin.
  • the method comprises delivering to a patient a nucleic acid encoding tyrosinase.
  • the method results in an increase in the ultraviolet absorption of the patient's skin.
  • Still other embodiments are directed to a method for reducing photodamage to a person's skin upon exposure to ultraviolet light.
  • the method results in the reduction of photodamage to the person's skin upon exposure to ultraviolet light.
  • Still other embodiments are directed to a method for treating xeroderma pigmentosum.
  • the method comprises delivering to a patient a nucleic acid encoding tyrosinase.
  • Still other embodiments are directed to a method for treating epidermolysis bullosa.
  • the method comprises delivering to a patient a nucleic acid encoding one or more of keratin 5, keratin 14, plectin, an integrin family member, laminin, a laminin subunit, collagen XVII, collagen VII or a biologically active fragment, variant, analogue or family-member thereof.
  • the method comprises delivering to a patient a nucleic acid encoding collagen type VII.
  • the method comprises delivering to a patient a nucleic acid encoding melanocortin 1 receptor.
  • Still other embodiments are directed to a method for treating xerosis.
  • the method comprises delivering to a patient a nucleic acid encoding a hyaluronan synthase.
  • the patient is diagnosed with atopic dermatitis.
  • the patient is diagnosed with ichthyosis.
  • Certain embodiments are directed to a method for treating a cosmetic condition.
  • Other embodiments are directed to a method for inducing tissue healing.
  • the method comprises delivering to a patient a nucleic acid encoding a hyaluronan synthase.
  • the cosmetic condition is a member of the group: wrinkles, sagging skin, thin skin, discoloration, and dry skin.
  • the patient has had cataract surgery.
  • the nucleic acid is synthetic RNA.
  • the method results in the amelioration of one or more of the patient's symptoms.
  • Other embodiments are directed to a method for treating an indication by delivering to a cell or a patient a nucleic acid encoding a protein or a peptide.
  • Still other embodiments are directed to a composition comprising a nucleic acid encoding a protein or a peptide.
  • Indications that can be treated using the methods and compositions of the present invention and proteins and peptides that can be encoded by compositions of the present invention are set forth in Table 2A and/or Table 2B, and are given by way of example, and not by way of limitation.
  • the indication is selected from Table 2A and/or Table 2B.
  • the protein or peptide is selected from Table 2A and/or Table 2B.
  • the indication and the protein or peptide are selected from the same row of Table 2A and/or Table 2B.
  • the protein of interest is a member of the group: UCP1, UCP2, and UCP3.
  • Other embodiments are directed to methods for inducing a cell to express a plurality of proteins of interest.
  • the proteins of interest include at least two members of the group: a lipase, UCP1, UCP2, and UCP3.
  • the proteins of interest include a lipase and a member of the group: UCP1, UCP2, and UCP3.
  • the protein is a gene-editing protein.
  • the gene-editing protein targets a gene that is at least partly responsible for a disease phenotype.
  • the gene-editing protein targets a gene that encodes a protein selected from Table 2A and/or Table 2B.
  • the gene-editing protein corrects or eliminates, either alone or in combination with one or more other molecules or gene-editing proteins, a mutation that is at least partly responsible for a disease phenotype.
  • the present invention contemplates the targeting of the precursor forms and/or mature forms and/or isoforms and/or mutants of any of the proteins disclosed in Table 2A and/or Table 2B and such proteins.
  • any of the precursor forms and/or mature forms and/or isoforms and/or mutants have enhanced secretion relative to the corresponding wild type proteins.
  • any of the precursor forms and/or mature forms and/or isoforms and/or mutants have altered half-lives (e.g. serum, plasma, intracellular)—for instance, longer or shorter half-lives. In some embodiments, this is relative to wild type.
  • Illustrative Indication Illustrative Protein/Peptide Acne Retinol Dehydrogenase 10 Aging Elastin sp
  • Additional illustrative targets of the present invention include the cosmetic targets listed in Table 6 of International Patent Publication No. WO 2013/151671, the contents of which are hereby incorporated by reference in their entirety.
  • the agents of the present invention are used in methods the effect the integumentary system of a human.
  • the present compositions and methods may be used to alter a biological and/or physiological process to, for example, reduce skin sagging, increase skin thickness, increase skin volume, reduce the number of wrinkles, the length of wrinkles and/or the depth of wrinkles, increase skin tightness, firmness, tone and/or elasticity, increase skin hydration and ability to retain moisture, water flow and osmotic balance, increase the levels of skin lipids; increase the extracellular matrix and/or adhesion and communication polypeptides; increase skin energy production; utilization and conservation; improve oxygen utilization; improve skin cell life; improve skin cell immunity defense, heat shock stress response, antioxidant defense capacity to neutralize free radicals, and/or toxic defense; improve the protection and recovery from ultraviolet rays; improve skin cell communication and skin cell innervations; improve cell cohesion/adhesion; improve calcium mineral and other mineral metabolism; improve cell turnover; and improve cell circadian rhythms.
  • the present compositions may be used in the treatment, control, or prevention of a disease, disorder and/or condition and/or may alter, modify or change the appearance of a member of the integumentary system of a subject suffering from a disease, disorder and/or condition such as, but not limited to, acne vulgaris, acne aestivalis, acne conglobata, acne cosmetic, acne fulminans, acne keloidalis nuchae, acne mechanica, acne medicamentosa, acne miliaris necrotica, acne necrotica, acne rosacea, actinic keratosis, acne vulgaris, acne aestivalis, acne conglobata, acne cosmetic, acne fulminans, acne keloidalis nuchae, acne mechanica, acne medicamentosa, acne miliaris necrotica, acne necrotica, acne rosacea, acute urticaria, allergic contact dermatitis, alopecia areata, angioedema, athlete's foot, atopic dermatiti
  • hidradenitis suppurativa hives, hyperhidrosis, hyperpigmentation, hypohidrotic ectodermal dysplasia, hypopigmentation, impetigo, ingrown hair, heat type urticara, ingrown toenail, infantile acne or neonatal acne, itch, irritant contact dermatitis, jock itch, keloid, keratosis pilaris, lichen planus, lichen sclerosus, lupus miliaris disseminatus faciei, melasma, moles, molluscum contagiosum, nail growth rate, nail health, neurodermatitis, nummular eczema, occupational acne, oil acne, onychomycosis, physical urticara, pilonidal cyst, pityriasis rosea, pityriasis versicolor, poison ivy, pomade acne, pseudofolliculitis barba
  • profilaggrin a protein which is converted to filaggrin
  • a protein of interest e.g. when treating ichthyosis vulgaris.
  • the protein of interest is any of the products of the genes psoriasis susceptibility 1 through 9 (PSORSI-PSORS9).
  • eczema e.g. atopic dermatitis, nummular eczema, dyshidrotic eczema, seborrheic dermatitis, irritant contact dermatitis, allergic contact dermatitis, dyshidrosis, venous eczema, dermatitis herpetiformis, neurodermatitis, autoeczematization and xerotic eczema
  • eczema e.g. atopic dermatitis, nummular eczema, dyshidrotic eczema, seborrheic dermatitis, irritant contact dermatitis, allergic contact dermatitis, dyshidrosis, venous eczema, dermatitis herpetiformis, neurodermatitis, autoeczematization and xerotic eczema
  • filaggrin three genetic variants, ovo-like 1 (OVOL1), actin-like 9 (ACT
  • Hives or urticaria, including, but not limited to, acute urticaria, chronic urticara and angioedema, physical urticara, pressure or delayed pressue urticara, cholinergic or stress uricara, cold type urticara, heat type urticara, solar type urticara, rare or water type urticara, vibratory angioedema, exercise-induced anaphylasis and dermatographism may be treated with the present compositions by, for example, targeting PLCG-2.
  • rosacea which includes, but is not limited to, erthematotelangiectatic rosacea, papulopustular rosacea, phymatous rosacea, and ocular rosacea.
  • CAMP cathelicidin antimicrobial peptide
  • SCTE stratum corneum tryptic enzyme
  • acne may include, but is not limited to, acneiform eruptions, acne aestivalis, acne conglobata, acne cosmetic, acne fulminans, acne keloidalis nuchae, acne mechanica, acne medicamentosa, acne miliaris necrotica, acne necrotica, acne rosacea, baby acne, blackheads, chloracne, excoriated acne, halogen acne, infantile acne or neonatal acne, lupus miliaris disseminatus faciei, occupational acne, oil acne, pomade acne, tar acne, tropical acne, tycoon's cap or acne necrotica miliaris, pseudofolliculitis barbae or acne keloidalis nuchae, and hidradenitis suppurativa.
  • the protein of interest may be one or more matrix metalloproteinases (MMP), e.g., matrix metalloproteinase-1 (MMP-1 or interstitial collagenase), matrix metalloproteinase-9 (MMP-9), and matrix metalloproteinase-13 (MMP-13).
  • MMP matrix metalloproteinases
  • MMP-1 or interstitial collagenase matrix metalloproteinase-1
  • MMP-9 matrix metalloproteinase-9
  • MMP-13 matrix metalloproteinase-13
  • vitiligo is treated with the present compositions, e.g. wherein the NLR family, pyrin domain containing 1 gene (NALP1) gene is targeted.
  • NLR family, pyrin domain containing 1 gene (NALP1) gene is targeted.
  • the present compositions find use in the treatment, control, or prevention of hyprohidrotic ectodermal dysplasia (HED), e.g. via the ectodysplasin A gene (EDA), receptor (EDAR), and receptor associated death domain (EDARADD).
  • HED hyprohidrotic ectodermal dysplasia
  • EDA ectodysplasin A gene
  • EDAR EDAR
  • EDARADD receptor associated death domain
  • the present compositions find use in the treatment, control, or prevention of balding, or hair thinning (e.g. male pattern baldness, or androgenetic alopecia (AGA)) and, optionally, one or more of the following may be the protein of interest: androgen receptor (AR), ectodysplasin A2 receptor (EDA2R) and lysophosphatidic acid receptor 6 (P2RY5).
  • AGA androgenetic alopecia
  • AR androgen receptor
  • EDA2R ectodysplasin A2 receptor
  • P2RY5 lysophosphatidic acid receptor 6
  • compositions also find use in methods of treatment, control, or prevention of scars and stretch marks (striae), e.g. via collagen, ribosomal s6 kinase, sectrected phosphoprotein 1 (also known as osteopontin), or transforming growth factor beta 3.
  • striae e.g. via collagen, ribosomal s6 kinase, sectrected phosphoprotein 1 (also known as osteopontin), or transforming growth factor beta 3.
  • Epidermodysplasia verruciformis also known as Lutz-Lewandowsky epidermodysplasia
  • a rare autosomal recessive genetic hereditary skin disorder may also be treated with compositions of the present invention, e.g. by targeted transmembrane channel-like 6 (EVER1) or transmembrane channellike 8 (EVER2) genes.
  • EVER1 transmembrane channel-like 6
  • EVER2 transmembrane channellike 8
  • skin sagging, thinning or wrinkling may be treated, controlled or prevented with present composition, e.g. by targeting one or more of the proteins of interest such as collagen, elastin, fibroblast growth factor 7, TIMP metallopeptidase inhibitors, matrix metallopeptidases, superoxide dismutase and other extracellular matrix proteins and proteoglycans.
  • proteins of interest such as collagen, elastin, fibroblast growth factor 7, TIMP metallopeptidase inhibitors, matrix metallopeptidases, superoxide dismutase and other extracellular matrix proteins and proteoglycans.
  • Further embodiments are used in tanning of the skin, such as via melanocyte-stimulating hormone and/or pro-opiomelanocortin.
  • the present compositions may be used for wound treatment.
  • methods of treating, controlling or preventing wounds with the present compositions comprises additional steps of, for example, cleaning the wound bed to facilitate wound healing and closure, including, but not limited to: debridement, sharp debridement (surgical removal of dead or infected tissue from a wound), optionally including chemical debriding agents, such as enzymes, to remove necrotic tissue; wound dressings to provide the wound with a moist, warm environment and to promote tissue repair and healing (e.g., wound dressings comprising hydrogels (e.g., AQUASORB; DUODERM), hydrocolloids (e.g., AQUACEL; COMFEEL), foams (e.g., LYOFOAM; SPYROSORB), and alginates (e.g., ALGISITE; CURASORB); administration of growth factors to stimulate cell division and proliferation and to promote wound healing e.g.
  • debridement sharp debridement (surgical removal of dead or in
  • a skin graft may be necessary to obtain coverage of clean, non-healing wounds (e.g., autologous skin grafts, cadaveric skin graft, bioengineered skin substitutes (e.g., APLIGRAF; DERMAGRAFT)).
  • clean, non-healing wounds e.g., autologous skin grafts, cadaveric skin graft, bioengineered skin substitutes (e.g., APLIGRAF; DERMAGRAFT)).
  • the nucleic acid drug described herein can be used in a variety of cosmetic/plastic surgery procedures, including, without limitation, a surgical procedure involving skin grafting and an aesthetic or cosmetic surgery (e.g. a facial plastic surgery procedure including, but not limited to blepharoplasty, rhinoplasty, rhytidectomy, genioplasty, facial implants, otoplasty, hair implantation, cleft lip and cleft palate repair, and/or a body plastic surgery procedure including but not limited to abdominoplasty, brachioplasty, thigh lift, breast reduction, breast augmentation, body contouring, liposuction, hand surgery).
  • a surgical procedure involving skin grafting and an aesthetic or cosmetic surgery
  • a facial plastic surgery procedure including, but not limited to blepharoplasty, rhinoplasty, rhytidectomy, genioplasty, facial implants, otoplasty, hair implantation, cleft lip and cleft palate repair
  • a body plastic surgery procedure including but not limited to abdominoplasty,
  • a variety of cancers are treated, controlled or prevented with the present compositions (e.g., colorectal cancer, gallbladder cancer, lung cancer, pancreatic cancer, and stomach cancer).
  • skin cancer is treated with the present compositions.
  • the skin cancer is one or more of actinic keratosis, basal cell carcinoma, melanoma, Kaposi's sarcoma, and squamous cell carcinoma.
  • the present compositions are used adjuvant to complete circumferential peripheral and deep margin assessment, Mohs surgery, radiation (e.g. external beam radiotherapy or brachytherapy), chemotherapy (including but not limited to topical chemotherapy, e.g.
  • compositions also find use in the treatment of various stages of cancers, including skin cancers (e.g. basal cell cancer (BCC), squamous cell cancer (SCC), and melanoma), such as a stage of the American Joint Committee on Cancer (AJCC) TNM system (e.g. one or more of TX, T0, Tis, T1, T1a, T1b, T2, T2A, T2B, T3, T3a, T3b, T4, T4a, T4b, NX, N0, N1, N2, N3, M0, M1a, M1b, M1c) and/or a staging system (e.g. Stage 0, Stage IA, Stage IB, Stage IIA, Stage IIB, Stage IIC, Stage IIIA, Stage IIIB, Stage IIIC, Stage IV).
  • BCC basal cell cancer
  • SCC squamous cell cancer
  • melanoma such as a stage of the American Joint Committee on Cancer (AJCC) TNM system
  • AJCC American
  • Illustrative cancers and/or tumors of the present invention include, but are not limited to, a basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx);
  • one or more rare diseases are treated, controlled or prevented with the present compositions, including, by way of illustration, Erythropoietic Protoporphyria, Hailey-Hailey Disease, Epidermolysis Bullosa (EB), Xeroderma Pigmentosum, Ehlers-Danlos Syndrome, Cutis Laxa, Protein C & Protein S Deficiency, Alport Syndrome, Striate Palmoplantar Keratoderma, Lethal Acantholytic EB, Pseudoxanthoma Elasticum (PXE), Ichthyosis Vulgaris, Pemphigus Vulgaris, and Basal Cell Nevus Syndrome.
  • Erythropoietic Protoporphyria Hailey-Hailey Disease
  • EB Epidermolysis Bullosa
  • Xeroderma Pigmentosum Ehlers-Danlos Syndrome
  • Cutis Laxa Protein C & Protein S Deficiency
  • Alport Syndrome Striate Palmoplant
  • the present compositions are used to treat, control or prevent one or more inflammatory diseases or conditions, such as inflammation, acute inflammation, chronic inflammation, respiratory disease, atherosclerosis, restenosis, asthma, allergic rhinitis, atopic dermatitis, septic shock, rheumatoid arthritis, inflammatory bowel disease, inflammatory pelvic disease, pain, ocular inflammatory disease, celiac disease, Leigh Syndrome, Glycerol Kinase Deficiency, Familial eosinophilia (FE), autosomal recessive spastic ataxia, laryngeal inflammatory disease; Tuberculosis, Chronic cholecystitis, Bronchiectasis, Silicosis and other pneumoconioses.
  • inflammatory diseases or conditions such as inflammation, acute inflammation, chronic inflammation, respiratory disease, atherosclerosis, restenosis, asthma, allergic rhinitis, atopic dermatitis, septic shock, rheumatoi
  • the present compositions are used to treat, control or prevent one or more autoimmune diseases or conditions, such as multiple sclerosis, diabetes mellitus, lupus, celiac disease, Crohn's disease, ulcerative colitis, Guillain-Barre syndrome, scleroderms, Goodpasture's syndrome, Wegener's granulomatosis, autoimmune epilepsy, Rasmussen's encephalitis, Primary biliary sclerosis, Sclerosing cholangitis, Autoimmune hepatitis, Addison's disease, Hashimoto's thyroiditis, Fibromyalgia, Menier's syndrome; transplantation rejection (e.g., prevention of allograft rejection) pernicious anemia, rheumatoid arthritis, systemic lupus erythematosus, dermatomyositis, Sjogren's syndrome, lupus erythematosus, multiple sclerosis, myasclerosis, mya
  • the present compositions are used to treat, control or prevent one or more neurologic diseases, including ADHD, AIDS—Neurological Complications, Absence of the Septum Pellucidum, Acquired Epileptiform Aphasia, Acute Disseminated Encephalomyelitis, Adrenoleukodystrophy, Agenesis of the Corpus Callosum, Agnosia, Aicardi Syndrome, Alexander Disease, Alpers' Disease, Alternating Hemiplegia, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Anencephaly, Aneurysm, Angelman Syndrome, Angiomatosis, Anoxia, Aphasia, Apraxia, Arachnoid Cysts, Arachnoiditis, Arnold-Chiari Malformation, Arteriovenous Malformation, Aspartame, Asperger Syndrome, Ataxia Telangiectasia, Ataxia, Attention Deficit-Hyperactivity Disorder, Autism, Autonomic Dysfunction, Back Pain, Barth Syndrome, Batten
  • the present compositions are used to treat one or more respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), bronchiectasis, allergic rhinitis, sinusitis, pulmonary vasoconstriction, inflammation, allergies, impeded respiration, respiratory distress syndrome, cystic fibrosis, pulmonary hypertension, pulmonary vasoconstriction, emphysema, Hantavirus pulmonary syndrome (HPS), Loeffler's syndrome, Goodpasture's syndrome, Pleurisy, pneumonitis, pulmonary edema, pulmonary fibrosis, Sarcoidosis, complications associated with respiratory syncitial virus infection, and other respiratory diseases.
  • respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), bronchiectasis, allergic rhinitis, sinusitis, pulmonary vasoconstriction, inflammation, allergies, impeded respiration, respiratory distress syndrome, cystic fibrosis, pulmonary hypertension, pulmonary va
  • the present compositions are used to treat, control or prevent cardiovascular disease, such as a disease or condition affecting the heart and vasculature, including but not limited to, coronary heart disease (CHD), cerebrovascular disease (CVD), aortic stenosis, peripheral vascular disease, atherosclerosis, arteriosclerosis, myocardial infarction (heart attack), cerebrovascular diseases (stroke), transient ischaemic attacks (TIA), angina (stable and unstable), atrial fibrillation, arrhythmia, vavular disease, and/or congestive heart failure.
  • cardiovascular disease such as a disease or condition affecting the heart and vasculature
  • CVD cerebrovascular disease
  • aortic stenosis aortic stenosis
  • peripheral vascular disease atherosclerosis
  • arteriosclerosis myocardial infarction
  • cerebrovascular diseases stroke
  • TIA transient ischaemic attacks
  • angina stable and unstable
  • atrial fibrillation arrhythmia, va
  • the present compositions are used to treat, control or prevent one or more metabolic-related disorders.
  • the present invention is useful for the treatment, controlling or prevention of diabetes, including Type 1 and Type 2 diabetes and diabetes associated with obesity.
  • the compositions and methods of the present invention are useful for the treatment or prevention of diabetes-related disorders, including without limitation diabetic nephropathy, hyperglycemia, impaired glucose tolerance, insulin resistance, obesity, lipid disorders, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels, high LDL levels, atherosclerosis and its sequelae, vascular restenosis, irritable bowel syndrome, inflammatory bowel disease, including Crohn's disease and ulcerative colitis, other inflammatory conditions, pancreatitis, abdominal obesity, neurodegenerative disease, retinopathy, neoplastic conditions, adipose cell tumors, adipose cell carcinomas, such as liposarcoma, prostate cancer and other cancers, including gastric, breast
  • a person has three or more of the following disorders: abdominal obesity, hypertriglyceridemia, low HDL cholesterol, high blood pressure, and high fasting plasma glucose), ovarian hyperandrogenism (polycystic ovary syndrome), and other disorders where insulin resistance is a component, such as sleep apnea.
  • the compositions and methods of the present invention are useful for the treatment, control, or prevention of obesity, including genetic or environmental, and obesity-related disorders.
  • the obesity-related disorders herein are associated with, caused by, or result from obesity.
  • obesity-related disorders include obesity, diabetes, overeating, binge eating, and bulimia, hypertension, elevated plasma insulin concentrations and insulin resistance, dyslipidemia, hyperlipidemia, endometrial, breast, prostate, kidney and colon cancer, osteoarthritis, obstructive sleep apnea, gallstones, heart disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovary disease, craniopharyngioma, Prader-Willi Syndrome, Frohlich's syndrome, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia.
  • obesity-related disorders are Metabolic Syndrome, insulin resistance syndrome, reproductive hormone abnormalities, sexual and reproductive dysfunction, such as impaired fertility, infertility, hypogonadism in males and hirsutism in females, fetal defects associated with maternal obesity, gastrointestinal motility disorders, such as obesity-related gastro-esophageal reflux, respiratory disorders, such as obesity-hypoventilation syndrome (Pickwickian syndrome), breathlessness, cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, lower back pain, gallbladder disease, hyperuricemia, gout, and kidney cancer, and increased anesthetic risk.
  • the compositions and methods of the present invention are also useful to treat Alzheimer's disease.
  • nucleic acids including liposomal formulations containing nucleic acids, when delivered in vivo, can accumulate in the liver and/or spleen. It has now been discovered that nucleic acids encoding proteins can modulate protein expression in the liver and spleen, and that nucleic acids used in this manner can constitute potent therapeutics for the treatment of liver and spleen diseases. Certain embodiments are therefore directed to a method for treating liver and/or spleen disease by delivering to a patient a nucleic acid encoding a protein of interest. Other embodiments are directed to a therapeutic composition comprising a nucleic acid encoding a protein of interest, for the treatment of liver and/or spleen disease.
  • Diseases and conditions of the liver and/or spleen that can be treated include, but are not limited to: hepatitis, alcohol-induced liver disease, drug-induced liver disease, Epstein Barr virus infection, adenovirus infection, cytomegalovirus infection, toxoplasmosis, Rocky Mountain spotted fever, non-alcoholic fatty liver disease, hemochromatosis, Wilson's Disease, Gilbert's Disease, and cancer of the liver and/or spleen.
  • the present compositions and methods relate to the treatment of type 1 diabetes, heart disease, including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, cystic fibrosis, sickle-cell anemia, thalassemia, Fanconi anemia, severe combined immunodeficiency, hereditary sensory neuropathy, xeroderma pigmentosum, Huntington's disease, muscular dystrophy, amyotrophic lateral sclerosis, Alzheimer's disease, cancer, and infectious diseases including hepatitis and HIV/AIDS.
  • type 1 diabetes including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, cystic fibrosis, sickle-cell anemia, thalassemia, Fanconi anemia, severe combined immunodeficiency, hereditary sensory neuropathy, xeroderma pigmentosum, Huntington's disease, muscular dystrophy, amyotrophic lateral sclerosis, Alzheimer's disease, cancer, and infectious diseases including hepatitis and HIV/AIDS
  • the present methods and compositions find use in targeting any of the proteins or in treatment of any of the diseases or disorders of Table 2B.
  • the present invention contemplates the targeting of the full-length and/or truncated forms of any of the proteins disclosed in Table 2B.
  • the present invention contemplates the targeting of the precursor forms and/or mature forms and/or isoforms of any of the proteins disclosed in Table 2B.
  • the present invention contemplates the targeting of a protein having about 60% (e.g. about 60%, or about 61%, or about 62%, or about 63%, or about 64%, or about 65%, or about 66%, or about 67%, or about 68%, or about 69%, or about 70%, or about 71%, or about 72%, or about 73%, or about 74%, or about 75%, or about 76%, or about 77%, or about 78%, or about 79%, or about 80%, or about 81%, or about 82%, or about 83%, or about 84%, or about 85%, or about 86%, or about 87%, or about 88%, or about 89%, or about 90%, or about 91%, or about 92%, or about 93%, or about 94%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99%) sequence identity with any of the protein sequences disclosed herein (e.g. in Table 2B).
  • the present invention contemplates the targeting of a protein comprising an amino acid sequence having one or more amino acid mutations relative to any of the protein sequences described herein (e.g. in Table 2B).
  • the present invention contemplates the targeting of a protein comprising an amino acid sequence having 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12 amino acid mutations relative to any of the protein sequences described herein (e.g. in Table 2B).
  • the one or more amino acid mutations may be independently selected from substitutions, insertions, deletions, and truncations.
  • the amino acid mutations are amino acid substitutions, and may include conservative and/or non-conservative substitutions. “Conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved.
  • the 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1) hydrophobic: Met, Ala, Val, Leu, Ile; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gln; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe.
  • “conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide.
  • glycine and proline may be substituted for one another based on their ability to disrupt ⁇ -helices.
  • non-conservative substitutions are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1) to (6) shown above.
  • the substitutions may also include non-classical amino acids (e.g. selenocysteine, pyrrolysine, N-formylmethionine ⁇ -alanine, GABA and ⁇ -Aminolevulinic acid, 4-aminobenzoic acid (PABA), D-isomers of the common amino acids, 2,4-diaminobutyric acid, ⁇ -amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, ⁇ -Abu, ⁇ -Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosme, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine,
  • Transthyretin his gene encodes transthyretin, one of the three prealbumins including alpha-1- (SEQ ID NOs: 637 and 638) antitrypsin, transthyretin and orosomucoid.
  • Transthyretin is a carrier protein; it Gene ID: 7276 transports thyroid hormones in the plasma and cerebrospinal fluid, and also transports retinol (vitamin A) in the plasma.
  • the protein consists of a tetramer of identical subunits.
  • This gene is regulated by (SEQ ID NO: 629 to 632) cytokines, suggesting that it may play a role in endothelium-dependent pathological Gene ID: 11082 disorders.
  • the transcript contains multiple polyadenylation and mRNA instability signals. Two transcript variants encoding different isoforms have been found for this gene
  • Parathyroid hormone PTH is secreted by the chief cells of the parathyroid glands. PTH plays an important P01270
  • Parathyroid hormone (SEQ ID NO: 508)
  • BMP-1 GeneSeq BMP1 belongs to the transforming growth factor-beta (TGFB) super- family.
  • Bone Accession P80618 morphogenic proteins induce cartilage and bone formation, play important role in WO8800205 nephrogesis, and play an important role in the development of many organs, P13497/BMP1_HUMAN including lung, heart, teeth, gut, skin, and particularly the kidney.
  • BMP-1 activity can Bone morphogenetic be determined using the following assays known in the art: Nat Genet. 2001 January; protein 1 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, (isoform BMP1-3) Issue 16, 10897-10902, Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev.
  • Bone Accession P80619 morphogenic protein induces bone formation.
  • BMP-2 activity can be determined WO8800205 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J P12643/BMP2_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902, Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594. Protein 2 Induction of Cartilage, Tissue and Bone Growth, and Diabetes. Infarction recovery. (SEQ ID NO: 170) Bone repair. Osteoporosis.
  • BMP-3 BMP-3 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone P12645
  • TGFB transforming growth factor-beta
  • BMP-2B GeneSeq BMP-2b belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession W24850 U.S. morphogenic protein induces bone formation. BMP-2b activity can be determined Pat. No. 5,631,142 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J P12644/BMP4_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; I Biol Cbcre, Vol. 274, Issue 16, 10897-10902, Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
  • TGFB transforming growth factor-beta
  • BMP-4 GeneSeq BMP-4 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession B02796 morphogenic protein induces bone formation. BMP-4 activity can be determined WO0020591 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J P12644/BMP4_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902, Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
  • BMP-5 GeneSeq BMP-5 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession B02797 morphogenic protein induces bone formation. BMP-5 activity can be determined WO0020591 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J P22003/BMP5_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902, Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
  • TGFB transforming growth factor-beta
  • BMP-6 GeneSeq BMP-6 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession R32904 U.S. morphogenic protein induces bone formation. BMP-6 activity can be determined Pat. No. 5,187,076 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J P22004/BMP6_HUMAN Biochem 1996 Apr.
  • OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone (SEQ ID NO: 579) morphogenic protein induces bone formation. OP-1 activity can be determined using the following assays known in the art: Nat Genet.
  • BMP7 Variant B OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone (SEQ ID NO: 580) morphogenic protein induces bone formation. OP-1 activity can be determined using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr.
  • OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily.
  • Bone (SEQ ID NO: 581) morphogenic protein induces bone formation.
  • OP-1 activity can be determined using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol.
  • OP-2 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone GeneSeq Accession morphogenic protein induces bone formation. OP-2 activity can be determined using R57973 WO9406399 the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J P34820/BMP8B_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol.
  • TGFB transforming growth factor-beta
  • GDF-1 The effect of GDF-1 on signaling can be assayed by treating Primary BAECs transferred with a construct called p3TP-Lux, containing a TGF-beta responsive promoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347).
  • TGFB transforming growth factor-beta
  • Bone Accession R86903 morphogenic protein induces bone formation.
  • BMP-9 activity can be determined WO9533830 using the following assays known in the art: Nat Genet.
  • BMP-10 GeneSeq BMP-10 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession R66202 morphogenic protein induces bone formation. BMP-10 activity can be determined WO9426893 using the following assays known in the art: Nat Genet.
  • BMP-12 GeneSeq BMP-12 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession R78734 morphogenic protein induces bone formation. BMP-12 activity can be determined WO9516035 using the following assays known in the art: Nat Genet.
  • BMP-15 GeneSeq BMP-15 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession W11261 morphogenic protein induces bone formation. BMP-15 activity can be determined W09636710 using the following assays known in the art: Nat Genet.
  • BMP-17 GeneSeq BMP-17 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone Accession Y17870 morphogenic protein induces bone formation. BMP-17 activity can be determined WO9929718 using the following assays known in the art: Nat Genet.
  • BMP-18 activity can be determined WO9929718 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J SEQ ID NO: 4 from U.S. Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902, Pat. No. 7,151,086 Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
  • inhibin beta A subunit joins the alpha subunit to form a pituitary FSH secretion Accession B02806 inhibitor.
  • Inhibin has been shown to regulate gonadal stromal cell proliferation WO0020591 negatively and to have tumor-suppressor activity.
  • serum levels of inhibin P05111/INHA_HUMAN have been shown to reflect the size of granulosa-cell tumors and can therefore be Inhibin alpha chain used as a marker for primary as well as recurrent disease.
  • Tumor suppressor activity (SEQ ID NO: 184) of inhibin can be determined using assays known in the art: Matzuk et al., Nature 1992 Nov. 26: 360 (6402); 313-9. Tumor suppression.
  • Inhibin beta GeneSeq The inhibin beta A subunit joins the alpha subunit to form a pituitary FSH secretion Accession H02808 inhibitor. Inhibin has been shown to regulate gonadal stromal cell proliferation WO0020591 negatively and to have tumour-suppressor activity.
  • inhibin P08476/INHBA_HUMAN serum levels of inhibin P08476/INHBA_HUMAN have been shown to reflect the size of granulosa-cell tumors and can therefore be Inhibin beta A chain used as a marker for primary as well as recurrent disease.
  • Tumor suppressor activity (SEQ ID NO: 185) of inhibin can be determined using assays known in the art: Matzuk et al., Nature P09529/INHBB_HUMAN 1992 Nov. 26: 360 (6402); 313-9. Tumor suppression.
  • Inhibin beta B chain (SEQ ID NO: 186) Cerberus Protein Cerebus is believed to be involved in the inhibition of BMP activity BMP activity, in the GeneSeq Accession presence of the antagonist Cerebus, can be determined using the following assays W86032 WO9849296 known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr. 1; O95813/CER1_HUMAN 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902, Apr. 16, 1999; and Cerberus Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
  • BMP Antagonist useful for (SEQ ID NO: 187) Osteosarcoma, abnormal bone growth.
  • Soluble BMP Receptor Soluble BMP receptor kinase protein-3 is involved in the binding of BMPs.
  • Soluble Kinase Protein-3 BMP receptor kinase protein-3 is useful as an antagonist for the inhibition of BMP GeneSeq Accession activity.
  • BMP activity, in the presence of the soluble antagonist BMP receptor kinase R95227 WO9614579 protein-3 can be determined using the following assays known in the art: Nat Genet. Q13873/BMPR2_HUMAN 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr.
  • BMP Antagonist useful for Osteosarcoma abnormal bone (SEQ ID NO: 188) growth.
  • BMP Processing Enzyme BMPs belong to the transforming growth factor-beta (TGFB) superfamily.
  • TGFB transforming growth factor-beta
  • Bone Furin GeneSeq Accession morphogenic protein induces bone formation.
  • BMP activity, in the presence of the W36099 WO9741250 Furin can be determined using the following assays known in the art: Nat Genet.
  • TGF-beta 1 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to Accession R29657 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII) WO9216228 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends P01137/TGFB1_HUMAN Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220). Transforming growth Activation of this heteromeric receptor complex occurs when TGF-beta.
  • TbetaRI binds to factor beta-1 TbetaRII, which then recruits and phosphorylates TbetaRI.
  • Activated TbetaRI propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995) PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341.
  • the effect of TGF betas on signaling can be assayed by treating Primary BAECs transfected with a construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347).
  • TGF-beta 2 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to Accession R39659 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII) EP542679 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends P61812/TGFB2_HUMAN Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220). Transforming growth Activation of this heteromeric receptor complex occurs when TGF-beta.
  • TbetaRI type I
  • TbetaRII type II
  • TbetaRI binds to factor beta-2 TbetaRII, which then recruits and phosphorylates TbetaRI.
  • Activated TbetaRI propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995) PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341.
  • the effect of TGF betas on signaling can be assayed by treating Primary BAECs transfected with a construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347).
  • ZTGF-beta 9 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to Accession Y70654 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII) WO0015798 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends SEQ ID NO: 2 of Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220). WO0015798 Activation of this heteromeric receptor complex occurs when TGF-beta.
  • TbetaRII binds to (SEQ ID NO: 192) TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995) PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341.
  • the effect of TGF betas on signaling can be assayed by treating Primary BAECs transfected with a construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347).
  • Anti-TGF beta family Members of the TGF-beta family of proteins initiate cell signaling by binding to antibodies GB2305921 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII) serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220). Activation of this heteromeric receptor complex occurs when TGF-beta. binds to TbetaRII, which then recruits and phosphorylates TbetaRI.
  • TbetaRI type I serine/threonine kinase receptors
  • TbetaRI then propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995) PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341.
  • the effect of TGF betas on signaling in the presence of an anti-TGF beta antibody can be assayed by treating Primary BAECs transfected with a construct called p3TP-Lux, containing a TGF-beta responsive promoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347).
  • TGF-betaRI protein II GeneSeq heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII) Accession Y70552 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends WO0012551 Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
  • Q14767/LTBP2_HUMAN Activation of this heteromeric receptor complex occurs when TGF-beta.
  • TbetaRII Latent-transforming TbetaRII
  • TbetaRI Activated TbetaRI then growth factor beta-binding propagates the signal to downstream targets
  • TGF (SEQ ID NO: 193) betas on signaling in the presence of a TGF beta binding protein, can be assayed by treating Primary BAECs transfected with a construct called p3TP-Lux, containing a TGF-beta responsivepromoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347). Useful for inhibiting tissue or tumor growth.
  • TGF-betaRI type I
  • TbetaRII type II
  • WO9741250 serine/threonine kinase receptors
  • TbetaRII binds to factor 5 TbetaRII, which then recruits and phosphorylates TbetaRI.
  • Activated TbetaRI propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995) PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341.
  • the effect of TGF betas on signaling can be assayed by treating Primary BAECs transfected with a construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature 370: 341-347).
  • BMP activity, in the presence of b57 protein, WO9837195 can be determined using the following assays known in the art: Nat Genet. 2001 January; SEQ ID NO: 2 of 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue WO9837195 16, 1089-10902, Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Deve. 10, 1580-1594.
  • Resistin GeneSeq This gene belongs to the family defined by mouse FIZZI and FIZZ3/Resistin genes. Accession W69293 The characteristic feature of this family is the C-terminal stretch of 10 cys residues WO0064920 with identical spacing. The mouse homolog of this protein is secreted by adipocytes, Q9HD89/RETN_HUMAN may be the hormone potentially linking obesity to type II diabetes.
  • Ability of resistin to Resistin influence type II diabetes can be determined using assays known in the art: Pontoglio (isoform 1) et al., J Clin Invest 1998 May 15; 101(10): 2215-22.
  • Type II diabetes and Syndrome (SEQ ID NO: 196) X.
  • Galectin-4 GeneSeq Galectins are a family of carbohydrate-binding proteins characterized by an affinity for Accession W11841 beta- galactoside containing glycoconjugates.
  • Ability of Galectin-4 polypeptides to WO9703190 bind lactose can be determined using assays known in the art: Wada, et al., J Biol P56470/LEG4_HUMAN Chem 1997 Feb. 28; 272(9): 6078-86. Lactose intolerance.
  • Galectin-4 (SEQ ID NO: 197) APM-I; ACRP- 30; ACPR30 gene is exclusively expressed in adipose tissue.
  • ACRP30 is thought to Famoxin GeneSeq increase fatty acid oxidation by muscle tissue.
  • Ability of ACRP30 polypeptides to Accession Y71035 influence obesity and fat oxidation can be determined using assays known in the art: W00026363 Fruebis et al., Proc Nat'l Acad Sci USA 2001 Feb. 13; 98(4): 2005-10.
  • ACRP30 is thought to Complement Component increase fatty acid oxidation by muscle tissue.
  • ACRP30 homologue Clq C GeneSeq Accession polypeptides to influence obesity and fat oxidation can be determined using assays B30234 WO0063376 known in the art: Fruebis et al., Proc Nat'l Acad Sci USA 2001 Feb. 13; 98(4): 2005-10. P02747/C1QC_HUMAN Obesity, Metabolic disorders, Lipid Metabolism; Hormone Secretion.
  • Complement C1q subcomponent subunit C SEQ ID NO: 199
  • Calpain-10a GeneSeq Calpain is believed to play a role in insulin secretion and insulin activity, and therefore Accession Y79567 may be useful in the treatment of type II diabetes.
  • Calpain-10 to influence WO0023603 type II diabetes can be determined using assays known in the art: Pontoglio et al., J Q9HC96/CAN10_HUMAN Clin Invest 1998 May 15; 101(10): 2215-22. Diabetes mellitus; Regulation of Insulin Calpain-10 secretory response; Insulin mediated glucose transport disorders.
  • Isoform A SEQ ID NO: 200
  • Calpain-10b GeneSeq Calpain is believed to play a role in insulin secretion and insulin activity, and therefore Accession Y79568 may be useful in the treatment of type II diabetes.
  • Calpain-10 ability of Calpain-10 to influence WO0023603 type II diabetes can be determined using assays known in the art: Pontoglio et al., J Q9HC96- Clin Invest 1998 May 15; 101(10): 2215-22. Diabetes mellitus; Regulation of Insulin 2/CAN10_HUMAN Isoform secretory response; Insulin mediated glucose transport disorders.
  • B of Calpain-10 (SEQ ID NO: 201) Calpain-10c GeneSeq Calpain is believed to play a role in insulin secretion and insulin activity, and therefore Accession Y79569 may be useful in the treatment of type II diabetes.
  • Calpain-10 ability of Calpain-10 to influence WO0023603 type II diabetes can be determined using assays known in the art: Pontoglio et al., J Q9HC96- Clin Invest 1998 May 15; 101(10): 2215-22. Diabetes mellitus; Regulation of Insulin 3/CAN10_HUMAN Isoform secretory response; Insulin mediated glucose transport disorders.
  • C of Calpain-10 SEQ ID NO: 202)
  • PDGF-D GeneSeq Vascular Endothelial Growth Factor.
  • Activity can be Y28594 WO9936079 determined using Apoptosis assays known in the art: Walczak et al. (1996) EMBOJ P48023/TNFL6_HUMAN 16: 5386-5397.
  • Chondro modulin-like Chondromodulin proteins are cartilage proteins thought to confer resistance to protein GeneSeq anglogeneis, and thus are useful as anti-angiogenic agents that may have utility in Accession Y71262 combating cancer.
  • Ability of Chondromodulin-like protein to inhibit vascularization can WO0029579 be determined using assays known in the art: Hirakie et al., J Biol Chem 1997 Dec. SEQ ID NO: 2 from 19; 272(51): 32419-26.
  • Antianglogenic agent; Osteoblast proliferation stimulator; WO0029579 prevents vascularization of cartilage tissue; Useful to treat cancer.
  • Patched GeneSeq Patched is a tumour-suppressor receptor for Sonic hedgehog (shh), which is a protein Accession W72969 U.S. that controls developmental patterning and growth.
  • Sohh Sonic hedgehog
  • Ability of soluble Patched to bind Pat. No. 5,837,538 to and inhibit the activities of shh can be determined using assays known in the art: Q13635/PTC1_HUMAN Stone et al., Nature 1996 Nov. 14; 384(6605): 129-34.
  • Receptor for Hedgehog cellular Protein patched homolog 1 proliferation signaling molecule This receptor is useful as a means of preventing (isoform L) cellular proliferation via the shh signaling path- way, thus useful for cancers.
  • Patched-2 GeneSeq Patched is a tumour-suppressor receptor for Sonic hedgehog (shh), which is a protein Accession Y43261 that controls developmental patterning and growth.
  • Ability of soluble Patched to bind WO9953058 to and inhibit the activities of shh can be determined using assays known in the art: Q9Y6C5/PTC2_HUMAN Stone et al., Nature 1996 Nov. 14; 384(6605): 129-34.
  • Receptor for Hedgehog cellular Protein patched homolog 2 proliferation signaling molecule This receptor is useful as a means of preventing (isoform 1) cellular proliferation via the shh signaling path- way, thus useful for cancers.
  • the maspin protein has tumour suppressing and (SEQ ID NO: 207) invasion sup- pressing activity.
  • Endostatin GeneSeq Endostatin is believed to inhibit effects of capillary endothelial cell proliferation.
  • the Accession B28399 inhibitory effects of endostatin can be assayed using assays disclosed by Cao et al. WO0064946 (1996) J. Biol. Chem. 271 29461-29467. Anti-angiogenic activity. Useful in the P39060/COIA1_HUMAN prevention and/or treatment of cancers.
  • Collagen alpha-1(XVIII) chain (isoform 1) (SEQ ID NO: 208) aFGF; FGF-1 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 Accession P94037 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein.
  • Antagonists may P05230/FGF1_HUMAN be useful as anti-cancer agents. Diabetes, Metabolic Disease, Obesity.
  • Fibroblast growth factor 1 (isoform 1) (SEQ ID NO: 209) bFGF; FGF-2 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 Accession R06685 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein.
  • Antagonists may P09038/FGF2_HUMAN be useful as anti-cancer agents.
  • Fibroblast growth factor 2 (isoform 1) (SEQ ID NO: 210) FGF-3; INT-2 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 Accession R07824 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth WO9503831 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may P11487/FGF3_HUMAN be useful as anti-cancer agents.
  • Fibroblast growth factor 3 (SEQ ID NO: 211) FGF-4; HST-1; HBGF-4 Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 GeneSeq Accession Cancer Res.
  • Antagonists may P12034/FGF5_HUMAN be useful as anti-cancer agents.
  • Fibroblast growth factor 5 isoform long
  • FGF-6 Heparin binding Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 secreted transforming Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein.
  • Antagonists may Accession R58555 be useful as anti-cancer agents.
  • EP613946 P10767/FGF6_HUMAN Fibroblast growth factor 6 (SEQ ID NO: 214) FGF-8 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 Accession R80783 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth WO9524928 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may P55075/FGF8_HUMAN be useful as anti-cancer agents.
  • Fibroblast growth factor 8 (isoform 8E) (SEQ ID NO: 215) FGF-9; Gila activating Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 factor GeneSeq Accession Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein.
  • P31371/FGF9_HUMAN Antagonists may be useful as anti-cancer agents.
  • Fibroblast growth factor 9 (SEQ ID NO: 216) FGF-12; Fibroblast growth Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 factor homologous factor-1 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth GeneSeq Accession and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may W06309 WO9635708 be useful as anti-cancer agents.
  • P61328/FGF12_HUMAN Fibroblast growth factor 12 (isoform 1) (SEQ ID NO: 217) FGF-19 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 Accession Y08582 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth WO9927100 and proliferation of cells, such as epithelial cells and keratinocytes. Chronic liver O95750/FGF19_HUMAN disease. Primary biliary cirrhosis. Bile acid-induced liver damage. Antagonists may be Fibroblast growth factor 19 useful as anti-cancer agents.
  • Fibroblast growth factor 18 (SEQ ID NO: 220) flt-3 ligand GeneSeq Stem Cell Progenitor Chemokine activities can be determined using assays known in Accession R67541 the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited EP627487 by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, P49771
  • Fms-related tyrosine kinase 3 ligand (isoform 1) (SEQ ID NO: 221)
  • VEGF-110 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession Y69417 determined using assays known in the art, such as those disclosed in International WO0013702 Publication No. WO0045835, for example.
  • Antagonists may be useful as anti- WO0013702 angiogenic agents, and may be applicable for cancer.
  • VEGF-121 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession B50432 determined using assays known in the art, such as those disclosed in International WO0071713 Publication No. WO0045835, for example. Promotion of growth and proliferation of SEQ ID NO: 2 from cells, such as vascular endothelial cells. Antagonists may be useful as anti- WO0071713 angiogenic agents, and may be applicable for cancer.
  • SEQ ID NO: 223 VEGF-138 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession Y43483 determined using assays known in the art, such as those disclosed in International WO9940197 Publication No. WO0045835, for example. Promotion of growth and proliferation of SEQ ID NO: 4 of cells, such as vascular endothelial cells. Antagonists may be useful as anti- WO99/40197 angiogenic agents, and may be applicable for cancer. (SEQ ID NO: 371) VEGF-145 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be Accession Y69413 determined using assays known in the art, such as those disclosed in International WO0013702 Publication No. WO0045835, for example.
  • SEQ ID NO: 4 Promotion of growth and proliferation of SEQ ID NO: 4 from cells, such as vascular endothelial cells.
  • Antagonists may be useful as anti- WO0013702 angiogenic agents, and may be applicable for cancer.
  • SEQ ID NO: 224 VEGF-162 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession Y43484 determined using assays known in the art, such as those disclosed in International W09940197 Publication No. WO0045835, for example.
  • Antagonists may be useful as anti- WO99/40197 angiogenic agents, and may be applicable for cancer.
  • VEGF-165 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession Y69414 determined using assays known in the art, such as those disclosed in International WO0013702 Publication No. WO0045835, for example. Promotion of growth and proliferation of SEQ ID NO: 6 from cells, such as vascular endothelial cells. Antagonists may be useful as anti- WO0013702 angiogenic agents, and may be applicable for cancer.
  • SEQ ID NO: 225 VEGF-182 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession Y43483 determined using assays known in the art, such as those disclosed in International W09940197 Publication No. WO0045835, for example. Promotion of growth and proliferation of SEQ ID NO: 6 of cells, such as vascular endothelial cells. Antagonists may be useful as anti- WO99/40197 angiogenic agents, and may be applicable for cancer. (SEQ ID NO: 373) VEGF-189 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be Accession Y69415 determined using assays known in the art, such as those disclosed in International WO0013702 Publication No. WO0045835, for example.
  • VEGF-206 GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession Y69416 determined using assays known in the art, such as those disclosed in International W00013702 Publication No. WO0045835, for example.
  • Antagonists may be useful as anti- WO0013702 angiogenic agents, and may be applicable for cancer.
  • VEGF-D GeneSeq Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be Accession W53240 determined using assays known in the art, such as those disclosed in International WO9807832 Publication No. WO0045835, for example. Promotion of growth and proliferation of O43915/VEGFD_HUMAN cells, such as vascular endothelial cells. Antagonists may be useful as anti- Vascular endothelial angiogenic agents, and may be applicable for cancer.
  • growth factor D (SEQ ID NO: 374) VEGF-E; VEGF-X Promotes the growth and/or proliferation of endothelial cells.
  • VEGF activity can be GeneSeq Accession determined using assays known in the art, such as those disclosed in International Y33679 WO9947677 Publication No. WO0045835, for example. Promotion of growth and proliferation of SEQ ID NO: 2 from cells, such as vascular endothelial cells. Antagonists may be useful as anti- WO9947677 angiogenic agents, and may be applicable for cancer.
  • VEGF Receptor (SEQ ID NO: 228) VEGF Receptor; KDR; flk- Receptor for VEGF polypeptides VEGF activity, in the presence of flk-1 polypeptides, 1 GeneSeq Accession can be determined using assays known in the art, such as those disclosed in W69679 WO9831794 International Publication No. WO0045835, for example. VEGF Receptor. Fusion P35968/VGFR2_HUMAN protein with the extracellular domain is useful as an anti-angiogenic agent. Vascular endothelial Antagonists may be useful in the promotion of angiogenesis.
  • VEGF Receptor Receptor for VEGF polypeptides VEGF activity in the presence of VEGF Receptor GeneSeq Accession polypeptides, can be determined using assays known in the art, such as those W47037 U.S. Pat. No. disclosed in International Publication No. WO0045835, for example.
  • VEGF Receptor. 5,712,380 Fusion protein with the extracellular domain is useful as an anti-angiogenic agent.
  • sVEGF-RI (FIG. 3) of Antagonists may be useful in the promotion of angiogenesis.
  • U.S. Pat. No. 5,712,380 SEQ ID NO: 442)
  • sVEGF-RII FIG.
  • flt-1 GeneSeq Accession Receptor for VEGF polypeptides VEGF activity, in the presence of flt-1 polypeptides, Y70751 WO0021560 can be determined using assays known in the art, such as those disclosed in P17948/VGFR1_HUMAN International Publication No. WO0045835, for example. VEGF Receptor.
  • Fusion Vascular endothelial protein with the extracellular domain is useful as an anti-angiogenic agent.
  • growth factor receptor 1 Antagonists may be useful in the promotion of angiogenesis.
  • (isoform 1) (SEQ ID NO: 230) VEGF R-3; flt-4 GeneSeq Receptor for VEGF polypeptides VEGF activity, in the presence of flt-4 polypeptides, Accession B29047 can be determined using assays known in the art, such as those disclosed in WO0058511 International Publication No. WO0045835, for example.
  • VEGF Receptor Fusion P35916/VGFR3_HUMAN protein with the extracellular domain is useful as an anti-angiogenic agent.
  • Vascular endothelial Antagonists may be useful in the promotion of angiogenesis.
  • growth factor receptor 3 isoform 1 (SEQ ID NO: 231)
  • Neuropilin-1 GeneSeq Vascular Endothelial Growth Factor VEGF activity can be determined using assays Accession Y06319 known in the art, such as those disclosed in International Publication No. WO9929858 WO0045835, for example.
  • Promotion of growth and proliferation of cells such as O14786/NRP1_HUMAN vascular endothelial cells.
  • Antagonists may be useful as anti-angiogenic agents, and Neuropilin-1 may be applicable for cancer.
  • isoform 1 (SEQ ID NO: 232)
  • Neuropilin-2 GeneSeq Vascular Endothelial Growth Factor VEGF activity can be determined using assays Accession Y03618 known in the art, such as those disclosed in International Publication No. WO9929858 WO0045835, for example. Promotion of growth and proliferation of cells, such as O60462/NRP2_HUMAN vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and Neuropilin-2 may be applicable for cancer.
  • (isoform A22) (SEQ ID NO: 233)
  • Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis.
  • High levels muscle troponin C may contribute to the difficulty encountered in revascularizing the ischemic GeneSeq Accession myocardium after cardiovascular injury.
  • Ability of soluble Troponins to inhibit W22597 W09730085 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci P02585/TNNC2_HUMAN USA 1999 Mar. 16; 96(6): 2645-50.
  • Anti-angiogenesis Troponin C, skeletal muscle (SEQ ID NO: 234) Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis.
  • High levels muscle troponin I may contribute to the difficulty encountered in revascularizing the ischemic GeneSeq Accession myocardium after cardiovascular injury.
  • soluble Troponins to inhibit W18054 W09730085 angiogenesis can be determined using assays known in the art. Proc Natl Acad Sci P48788/TNNI2_HUMAN USA 1999 Mar. 16; 96(6): 2645-50.
  • Anti-angiogenesis Troponin I fast skeletal muscle (isoform 1) (SEQ ID NO: 235) Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels muscle troponin T may contribute to the difficulty encountered in revascularizing the ischemic GeneSeq Accession myocardium after cardiovascular injury.
  • Ability of soluble Troponins to inhibit W22599 W09730085 angiogenesis can be determined using assays known in the art:.
  • Anti-angiogenesis WO9719955 (SEQ ID NO: 237) myofibrillar protein Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels troponin I GeneSeq may contribute to the difficulty encountered in revascularizing the ischemic Accession W18054 myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit WO9719955 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci SEQ ID NO: 3 of USA 1999 Mar. 16; 96(6): 2645-50.
  • Anti-angiogenesis WO9719955 (SEQ ID NO: 237) Troponin peptides Troponins are contractile proteins that are thought to inhibit angiogenesis.
  • GeneSeq Accessions may contribute to the difficulty encountered in revascularizing the ischemic Y29581, Y29582, Y29583, myocardium after cardiovascular injury.
  • Ability of soluble Troponins to inhibit Y29584, Y29585, and angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci Y29586 WO9933874 USA 1999 Mar. 16; 96(6): 2645-50.
  • Anti-angiogenesis Wildtype troponins provided as: Human fast twitch skeletal muscle troponin C GeneSeq Accession W22597 W09730085 P02585/TNNC2_HUMAN Troponin C, skeletal muscle (SEQ ID NO: 234) Human fast twitch skeletal muscle troponin I GeneSeq Accession W18054 W09730085 P48788/TNNI2_HUMAN Troponin I, fast skeletal muscle (isoform 1) (SEQ ID NO: 235) Human fast twitch skeletal muscle troponin T GeneSeq Accession W22599 W09730085 SEQ ID NO: 3 of WO9730085 (SEQ ID NO: 236) fragment.
  • High levels muscle Troponin subunit C may contribute to the difficulty encountered in revascularizing the ischemic GeneSeq Accession myocardium after cardiovascular injury.
  • Ability of soluble Troponins to inhibit B00134 WO0054770 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci SEQ ID NO: 1 of USA 1999 Mar. 16; 96(6): 2645-50.
  • Anti-angiogenesis WO0054770 (SEQ ID NO: 375) Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis.
  • High levels muscle Troponin subunit I may contribute to the difficulty encountered in revascularizing the ischemic Protein GeneSeq myocardium after cardiovascular injury.
  • soluble Troponins to inhibit Accession B00135 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci WO0054770 USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis SEQ ID NO: 2 of WO0054770 (SEQ ID NO: 376) Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels muscle Troponin subunit T may contribute to the difficulty encountered in revascularizing the ischemic GeneSeq Accession myocardium after cardiovascular injury.
  • Ability of soluble Troponins to inhibit B00136 WO0054770 angiogenesis can be determined using assays known in the art:.
  • PAI-1 PAIs are believed to play a role in cancer, and cardiovascular disease and blood- GeneSeq Accession clotting disorders.
  • Methods that measure plasminogen activator inhibitor (PAI) activity R08411 WO9013648 are known in the art, for example, assay the ability of PAI to inhibit tissue P05121/PAI1_HUMAN plasminogen activator (tPA) or urokinase (uPA): J Biochem Biophys Methods 2000 Plasminogen activator Sep.
  • inhibitor 1 measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad (isoform 1) Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders. (SEQ ID NO: 238) Plasminogen Activator PAIs are believed to play a role in cancer, and cardiovascular disease and blood- Inhibitor-2; PAI-2 clotting disorders.
  • PAI plasminogen activator inhibitor
  • tPA tissue P94160 DE3722673 plasminogen activator
  • uPA urokinase
  • Methods that Plasminogen activator measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad inhibitor 2 Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders.
  • PAI-2 PAIs are believed to play a role in cancer, and cardiovascular disease and blood- GeneSeq Accession clotting disorders.
  • Methods that measure plasminogen activator inhibitor (PAI) activity R10921 WO9102057 are known in the art, for example, assay the ability of PAI to inhibit tissue P05120/PAI2_HUMAN plasminogen activator (tPA) or urokinase (uPA): J Biochem Biophys Methods 2000 Plasminogen activator Sep. 11; 45(2): 127-40, Breast Cancer Res Treat 1996; 41(2): 141-6.
  • inhibitor 2 measure anti- angiogenesis activity
  • Proc Natl Acad SEQ ID NO: 239) Sci USA 1999 Mar. 16; 96(6): 2645-50.
  • Anti-angiogenesis ; blood-clotting disorders.
  • Human PAI-1 mutants PAIs are believed to play a role in cancer, and cardiovascular disease and blood- GeneSeq Accessions clotting disorders.
  • plasminogen activator inhibitor (PAI) activity R11755, R11756, R11757
  • Methods that R11763 WO9105048 measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad Wildtype PAI-1 is provided Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders.
  • Plasminogen activator inhibitor 1 (isoform 1) (SEQ ID NO: 238) CXCR3; CXC GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession Y79372 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO0018431 Members of this family are involved in a similarly diverse range of pathologies P49682
  • Chemokine activities can be determined (SEQ ID NO: 240) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Soluble CXCR3 polypeptides may be useful for inhibiting chemokine activities and viral infection.
  • Modified Rantes GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W38129 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO9737005 Members of this family are involved in a similarly diverse range of pathologies Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the herein as chemokines exert their effects by acting on a family of seven transmembrane G- P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-C motif chemokine 5 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders.
  • RANTES GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession Y05299 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • EP905240 Members of this family are involved in a similarly diverse range of pathologies P13501/CCL5_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 5 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 241) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • MCP-Ia GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession R73914 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO9509232 Members of this family are involved in a similarly diverse range of pathologies MCP-1 provided as including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P13500/CCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 2 protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological Accession Y26176 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9929728 Members of this family are involved in a similarly diverse range of pathologies MCP-1 provided as including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P13500/CCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 2 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 337) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • MCP-I receptor GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession R79165 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9519436 Members of this family are involved in a similarly diverse range of pathologies MCP-IA including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9519436 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 446) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined MCP-1B using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: SEQ ID NO: 4 of Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. WO9519436 Humana Press Inc., Totowa, NJ. Soluble MCP-1 Receptor polypep- tides may be (SEQ ID NO: 447) useful for inhibiting chemokine activities and viral infection.
  • MCP-3 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession R73915 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W09509232 Members of this family are involved in a similarly diverse range of pathologies P80098/CCL7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 7 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 336) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • MCP-4 receptor GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W56689 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9809171 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9809171 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 378) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Soluble MCP-4 Receptor polypep- tides may be useful for inhibiting chemokine activities and viral infection.
  • RANTES receptor Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W29588 U.S. Pat. No.
  • chemokines exert their effects by acting on a family of seven transmembrane G- No. 5,652,133 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 379) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • CCR5 variant GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W88238 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9854317 Members of this family are involved in a similarly diverse range of pathologies Variants of wildtype CCR5 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The which has the sequence chemokines exert their effects by acting on a family of seven transmembrane G- of: protein coupled receptors.
  • Chemokine activities can be determined C-C chemokine receptor using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: type 5 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. (SEQ ID NO: 448) Humana Press Inc., Totowa, NJ. Soluble CCR5 polypeptides may be useful for inhibiting chemokine activities and viral infection.
  • CCR7 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession B50859 U.S. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Pat. No. 6,153,441 Members of this family are involved in a similarly diverse range of pathologies P32248/CCR7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G- type 7 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 243) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Soluble CCR7 polypeptides may be useful for inhibiting chemokine activities and viral infection.
  • CXC3 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W23345 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9727299 Members of this family are involved in a similarly diverse range of pathologies P78423/X3CL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Fractalkine chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 244) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Eotaxin GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W10099 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO9700960 Members of this family are involved in a similarly diverse range of pathologies P51671/CCL11_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Eotaxin chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 245) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders.
  • Neurotactin GeneSeq Neurotactin may play a role in chemotactic leukocyte migration and brain Accessions Y77537, inflammation processes.
  • Chemotactic leukocyte migration assays are known in the W34307, Y53259, and, art, for example: J. Immunol. Methods 33, ((1980)); Nature 1997 Jun.
  • chemokines exert their effects by acting on a family of seven transmembrane G- No. 6,153,441 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 246) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders.
  • Lymphotactin GeneSeq are a family of related small, secreted proteins involved in biological Accession B50052 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO0073320 Members of this family are involved in a similarly diverse range of pathologies P47992/XCL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Lymphotactin chemokines exert their effects by acting on a family of seven transmembrane G. (SEQ ID NO: 247) Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I.
  • MIP-3 alpha GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W44398 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO9801557 Members of this family are involved in a similarly diverse range of pathologies P78556/CCL20_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 20 chemokines exert their effects by acting on a family of seven transmembrane G.
  • Chemokine activities can be determined using assays known in the art: Methods in (SEQ ID NO: 248) Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders.
  • MIP-3 beta GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W44399 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9801557 Members of this family are involved in a similarly diverse range of pathologies Q99731/CCL19_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 19 chemokines exert their effects by acting on a family of seven transmembrane G. (SEQ ID NO: 249) Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders.
  • Chemokines are a family of related small, secreted proteins involved in biological Accession R70798 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO2006135382 Members of this family are involved in a similarly diverse range of pathologies (SEQ ID NO: 457) including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The chemokines exert their effects by acting on a family of seven transmembrane G. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C.
  • Stem Cell Inhibitory Factor Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • R11553 WO9104274 Members of this family are involved in a similarly diverse range of pathologies SCIF in Table I of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9104274 chemokines exert their effects by acting on a family of seven transmembrane G.
  • Chemokine activities can be determined using assays known in the art: Methods in SCIF in Table II of Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, WO9104274 T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. (SEQ ID NO: 381) Hematopoietic growth factors. Thrombopoietin GeneSeq Thrombopoietin is involved in the regulation of the growth and differentiation of Accession R79905 megakaryocytes and preceptors thereof.
  • Thrombopoietin can be assayed to WO9521920 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol P40225
  • isoform 1 (SEQ ID NO: 250) c-kit ligand; SCF; Mast cell C-kit ligan is thought to stimulate the proliferation of mast cells, and is able to growth factor; MGF; augment the proliferation of both myeloid and lymphoid hematopoietic progenitors in Fibrosarcoma- derived bone marrow culture.
  • C-kit ligand is also though to act synergistically with other stem cell factor GeneSeq cytokines. Chemokine activities can be determined using assays known in the art: Accession Y53284, Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I.
  • Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays factor GeneSeq Accession known in the art, such as those disclosed in International Publication No. B48653 WO0066736 WO0045835, for example. Promotion of growth and proliferation of cells, such as PDGF-A vascular endothelial cells.
  • Antagonists may be useful as anti-angiogenic agents, and P04085/PDGFA_HUMAN may be applicable for cancer.
  • Platelet-derived growth factor subunit A Isoform long
  • PDGF-B P01127/PDGFB_HUMAN
  • Platelet-derived growth factor subunit B isoform 1
  • Melanoma inhibiting Melanoma inhibiting protein has melanoma-inhibiting activity and can be used to treat protein GeneSeq cancer (melanoma, glioblastoma, neuroblastoma, small cell lung cancer, Accession R69811 neuroectodermal tumors) or as an immunosuppressant (it inhibits IL-2 or WO9503328 phytohaemagglutinin induced proliferation of peripheral blood lymphocytes.
  • Tumor (SEQ ID NO: 458) suppressor activity of melanoma inhibiting protein can be determined using assays known in the art: Matzuk et al., Nature 1992 Nov. 26; 360(6402): 313-9. Cancer; melanoma Glioma-derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays factor GeneSeq Accession known in the art, such as those disclosed in International Publication No. R08120 EP399816 WO0045835, for example. Promotion of growth and proliferation of cells, such as vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and may be applicable for cancer.
  • Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays factor precursor A known in the art, such as those disclosed in International Publication No. GeneSeq Accession WO0045835, for example. Promotion of growth and proliferation of cells, such as R84759 EP682110 vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and PDGF-A precursor (variant may be applicable for cancer. D1) (SEQ ID NO: 382) PDGF-A precursor (variant 13-1) (SEQ ID NO: 383) Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays factor precursor B known in the art, such as those disclosed in International Publication No.
  • Wildtype PDGF-B provided as: PDGF-B P01127/PDGFB_HUMAN Platelet-derived growth factor subunit B (isoform 1) (SEQ ID NO: 258) Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays factor Bvsis GeneSeq known in the art, such as those disclosed in International Publication No. Accession P80595 and WO0045835, for example.
  • Antagonists may be useful as anti-angiogenic agents, and FIG. 1 of EP282317 may be applicable for cancer.
  • SEQ ID NO: 384 Placental Growth Factor Vascular Endothelial Growth Factor VEGF activity can be determined using assays GeneSeq Accessions known in the art, such as those disclosed in International Publication No. R23059 and R23060 WO0045835, for example.
  • Antagonists may be useful as anti-angiogenic agents, and P49763-2/PLGF_HUMAN may be applicable for cancer.
  • Isoform PIGF-1 of Placenta growth factor (isoform PIGF-1) (SEQ ID NO: 252) Placental Growth Factor-2 Vascular Endothelial Growth Factor VEGF activity can be determined using assays GeneSeq Accession known in the art, such as those disclosed in International Publication No. Y08289 DE19748734 WO0045835, for example. Promotion of growth and proliferation of cells, such as P49763-3/PLGF_HUMAN vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and Isoform PIGF-2 of may be applicable for cancer.
  • Placenta growth factor (isoform PIGF-2) (SEQ ID NO: 253)
  • Thrombopoietin Thrombopoietin is involved in the regulation of the growth and differentiation of derivative1 GeneSeq megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Accession Y77244 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Thrombopoietin is involved in the regulation of the growth and differentiation of derivative2 GeneSeq megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Accession Y77255 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 e.g. Table 3 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Thrombopoietin is involved in the regulation of the growth and differentiation of 3 GeneSeq Accession megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Y77262 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Thrombopoietin is involved in the regulation of the growth and differentiation of 4 GeneSeq Accession megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Y77267 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Thrombopoietin is involved in the regulation of the growth and differentiation of 5 GeneSeq Accession megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Y77246 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Thrombopoietin is involved in the regulation of the growth and differentiation of 6 GeneSeq Accession megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Y77253 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Thrombopoietin is involved in the regulation of the growth and differentiation of 7 GeneSeq Accession megakaryocytes and preceptors thereof.
  • Thrombopoietin (TPO) can be assayed to Y77256 determine regulation of growth and differentiation of megakaryocytes.
  • Mol Cell Biol WO0000612 e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within. Wildtype thrombopoietin Thrombocytopenia, cancer.
  • Fractalkine GeneSeq Fractalkine is believed to play a role in chemotactic leukocyte migration and Accession Y53255 U.S. neurological disorders. Fractalkine activity can be determined using Chemotactic Pat. No. 6,043,086 leukocyte migration assays known in the art, for example: J. Immunol. Methods 33, P78423/X3CL1_HUMAN ((1980)); Nature 1997 Jun. 5; 387(6633): 611-7. Immune disorders.
  • Fractalkine (SEQ ID NO: 244) CXC3 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W23345 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO9757599 Members of this family are involved in a similarly diverse range of pathologies P78423/X3CL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Fractalkine chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 244) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders.
  • CCR7 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession B50859 U.S. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Pat. No.
  • C-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G- type 7 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 243) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Trk tyrosine Accession R47100 kinase activation assays known in the art can be used to assay for neurotrophin WO9325684 activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
  • Trk tyrosine GeneSeq Accession kinase activation assays known in the art can be used to assay for neurotrophin R47101 WO9325684 activity, for example, Proc Natl Acad Sci USA 2001 Mar.
  • Wildtype neurotrophin Neurological disorders, cancer provided as: P34130/NTF4_HUMAN Neurotrophin-4 (SEQ ID NO: 256) Neurotrophin- 4b Neurotrophins regulate neuronal cell survival and synaptic plasticity.
  • tyrosine kinases GeneSeq Accession Trk tyrosine kinase activation assays known in the art can be used to assay for R47102 WO9325684 neurotrophin activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
  • P34130/NTF4_HUMAN Neurological disorders cancer Neurotrophin-4 (SEQ ID NO: 256) Neurotrophin- 4c Neurotrophins regulate neuronal cell survival and synaptic plasticity.
  • tyrosine kinases GeneSeq Accession Trk tyrosine kinase activation assays known in the art can be used to assay for R47103 WO9325684 neurotrophin activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
  • P34130/NTF4_HUMAN Neurological disorders cancer Neurotrophin-4 (SEQ ID NO: 256) Neurotrophin- 4d Neurotrophins regulate neuronal cell survival and synaptic plasticity.
  • tyrosine kinases GeneSeq Accession Trk tyrosine kinase activation assays known in the art can be used to assay for R47102 WO9325684 neurotrophin activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
  • P34130/NTF4_HUMAN Neurological disorders, cancer Neurotrophin-4 (SEQ ID NO: 256) Platelet-Derived Growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays Factor A chain GeneSeq known in the art, such as those disclosed in International Publication No. Accession R38918 U.S. W00045835, for example.
  • Antagonists may P04085/PDGFA_HUMAN be useful as anti-angiogenic agents, and may be applicable for cancer Platelet-derived growth factor subunit A (Isoform long) (SEQ ID NO: 257) Platelet-Derived Growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays Factor B chain GeneSeq known in the art, such as those disclosed in International Publication No. Accession R38919 U.S. W00045835, for example. Promotion of growth and proliferation of cells, such as Pat. No.
  • Antagonists may P01127/PDGFB_HUMAN be useful as anti-angiogenic agents, and may be applicable for cancer Platelet-derived growth factor subunit B (isoform 1) (SEQ ID NO: 258) Stromal Derived Factor- 1 Stromal Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 alpha GeneSeq Accession Cancer Res.
  • Immune coil Accession R78691 proliferation and suppression of apoptosis by prolactin can be assayed by methods WO9521625 well-known in the art, for example, Buckley, A R and Buckley D J, Ann NY Acad Sci P01236/PRL_HUMAN 2000; 917: 522-33, and within. Reproductive system disorders, cancer.
  • Prolactin SEQ ID NO: 262
  • Prolactin2 GeneSeq Prolactin is involved in immune cell proliferation and apoptosis.
  • Immune coil Accession Y31764 U.S. proliferation and suppression of apoptosis by prolactin can be assayed by methods Pat. No.
  • P01215/GLHA_HUMAN Glycoprotein hormones alpha chain (SEQ ID NO: 263) Follicle stimulating FSH stimulates secretion of interleukin-1 by cells isolated from women in the follicular hormone Beta subunit phase FSH activities can be determined using assays known in the art; J Gend Specif GeneSeq Accession Med 1999 November-December; 2(6): 30-4; Mol Cell Endocrinol. 1997 Nov. 15; 134(2): 109-18. Y54161 EP974359 Reproductive system disorders, cancer.
  • P01225/FSHB_HUMAN Follitropin subunit beta (SEQ ID NO: 264)
  • Substance P tachykinin
  • Substance P is associated with immunoregulation.
  • Oxytocin (Neurophysin I) Oxytocin is involved in the induction of prostaglandin (E2) release as well as an GeneSeq Accession increased amount of calcium release by smooth muscle cells.
  • Oxytocin and B24085 and B24086 prostaglandin E(2) release and Ocytocin (Ca2+) increase can be assayed by WO0053755 methods well-known in the art, for example, Pavan et al., AM J Obset Gynecol 2000 P01178/NEU1_HUMAN July; 183(1): 76-82 and Holda et al., Cell Calcium 1996 July; 20(1): 43 51.
  • Vasopressin Neurostyrene-like fibroblasts
  • fibroblasts fibroblasts
  • Vasopressin Neurosin Vasopressinis believed to have a direct antidiuretic action on the kidney, and it is II
  • GeneSeq Accession thought to cause vasoconstriction of the peripheral vessels.
  • Vasopressin activity can B24085 and B24086 be determined using assays known in the art, for example, Endocr Regul 1996 March; WO0053755 30(I): 13-17.
  • IL-1 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, P60326 EP165654 monocytes, and macrophages.
  • Known functions include stimulating proliferation of IL-1 alpha immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P01583
  • Interleukin activity Interleukin-1 alpha can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 269) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, IL-1 beta D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
  • IL-1 mature GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R14855 monocytes, and macrophages.
  • Known functions include stimulating proliferation of EP456332 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis (mature truncated form of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity wherein the precursor is can be determined using assays known in the art: Matthews et al., in Lymphokines cleaved between amino and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, acids 116-117) D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20. (SEQ ID NO: 386) inflammatory disorders immunologic disorders, cancer IL-1 beta GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession Y08322 monocytes, and macrophages.
  • Interleukin activity Interleukin-1 beta can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 267) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
  • inflammatory disorders immunologic disorders cancer IL-3 variants GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession P80382, monocytes, and macrophages.
  • Known functions include stimulating proliferation of P80383, P80384, and immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P80381 WO8806161 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Variants of wildtype IL-3 can be determined using assays known in the art: Matthews et al., in Lymphokines which has the sequence: and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, P08700
  • Interleukin-3 inflammatory disorders immunologic disorders, cancer SEQ ID NO: 449)
  • IL-4 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, P70615 WO8702990 monocytes, and macrophages.
  • Known functions include stimulating proliferation of P05112/IL4_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Interleukin-4 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 268) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132, 287-295.
  • inflammatory disorders immunologic disorders, cancer IL-4 muteins GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession W52151 monocytes, and macrophages.
  • Known functions include stimulating proliferation of W52152 W52153 W52154 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W52155 W52156 W52157 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity W52158 W52159 W52160 can be determined using assays known in the art: Matthews et al., in Lymphokines W52161 W52162 W52163 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, W52164 and W52165 D.C. 1987, pp.
  • inflammatory disorders immunologic disorders, cancer Variants of wildtype IL-4 which has the sequence: P05112/IL4_HUMAN Interleukin-4 (isoform 1) (SEQ ID NO: 268) IL-1 alpha GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession P90108 monocytes, and macrophages.
  • Interleukin activity Interleukin-1 alpha can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 269) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
  • inflammatory disorders immunologic disorders cancer IL-3 variants GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R38561, monocytes, and macrophages.
  • Known functions include stimulating proliferation of R38562, R38563, R38564, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis R38565, R38566, R38567, of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity R38568, R38569, R38570 can be determined using assays known in the art: Matthews et al., in Lymphokines R38571, and R38572 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, WO9307171 D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
  • IL-6 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, R45717 and R45718 monocytes, and macrophages.
  • Known functions include stimulating proliferation of WO9402512 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P05231/IL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-6 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 270) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16. inflammatory disorders immunologic disorders, cancer. Obesity. Metabolic Disease. Diabetes. IL-13 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, R48624 WO9404680 monocytes, and macrophages.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Boutelier et al (1995) J. Immunol. Methods 181, 29.
  • cancer IL-4 mutein GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R47182 monocytes, and macrophages.
  • Known functions include stimulating proliferation of DE4137333 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Variants of wildtype IL-4 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity which has the sequence: can be determined using assays known in the art: Matthews et al., in Lymphokines P05112/IL4_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132, (isoform 1) 287-295.
  • inflammatory disorders immunologic disorders, cancer SEQ ID NO: 268)
  • IL-4 mutein Y124X Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R47183 DE4137333 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Variants of wildtype IL-4 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity which has the sequence: can be determined using assays known in the art: Matthews et al., in Lymphokines P05112/IL4_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Interleukin-4 D.C. 1987, pp.
  • inflammatory disorders immunologic disorders, cancer SEQ ID NO: 268)
  • IL-4 mutein Y124G Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R47184 DE4137333 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Variants of wildtype IL-4 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity which has the sequence: can be determined using assays known in the art: Matthews et al., in Lymphokines P05112/IL4_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132, (isoform 1) 287-295. inflammatory disorders immunologic disorders, cancer (SEQ ID NO: 268) Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, (precursor) GeneSeq monocytes, and macrophages.
  • Interleukin activity P22301/IL10_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-10 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (precursor form is D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med.
  • Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R42642 WO9318783-A immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis SEQ ID NO: 3 of of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity WO9318783-A can be determined using assays known in the art: Matthews et al., in Lymphokines (mature IL-10) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 273) D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
  • inflammatory disorders immunologic disorders, cancer Human interleukin-1 beta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, precursor. GeneSeq monocytes, and macrophages.
  • Interleukin activity P01584/IL1B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-1 beta and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 274) D.C. 1987, pp.
  • inflammatory disorders immunologic disorders cancer Interleukin- 1alpha Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R45364 EP578278 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P01583
  • Interleukin activity Interleukin-1 alpha can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 269) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders, cancer Human interleukin-3 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, variant GeneSeq monocytes, and macrophages.
  • Interleukin activity Variants of wildtype IL-3 can be determined using assays known in the art: Matthews et al., in Lymphokines which has the sequence: and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, P08700
  • Interleukin-3 inflammatory disorders immunologic disorders, cancer (SEQ ID NO: 449)
  • IL-1i fragments GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R35484 and monocytes, and macrophages.
  • Known functions include stimulating proliferation of R35485 EP541920 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
  • inflammatory disorders immunologic disorders, cancer IL-1 inhibitor (IL-li) Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • R35486 and R35484 immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis EPS541920 of neutrophils and T lymphocytes and/or inhibition of interferons.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) inflammatory disorders immunologic disorders, cancer ICE 22 kD subunit.
  • Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R33780 EP533350 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis SEQ ID NO: 16 of of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity EP533350 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 450) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer ICE 20 kD subunit. Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R33781 EP533350 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis SEQ ID NO: 17 of of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity EP533350 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 451) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • cancer ICE 10 kD subunit Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R33782 EP533350 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis SEQ ID NO: 18 of of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity EP533350 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 452) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders, cancer Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, (precursor) GeneSeq monocytes, and macrophages.
  • Interleukin activity P22301/IL10_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-10 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (precursor form is D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med.
  • Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R42642 WO9318783 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis SEQ ID NO: 3 of of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity WO9318783-A can be determined using assays known in the art: Matthews et al., in Lymphokines (mature IL-10) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 273) D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
  • inflammatory disorders immunologic disorders, cancer Human Interleukin-1 beta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, precursor GeneSeq monocytes, and macrophages.
  • Interleukin activity P01584/IL1B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-1 beta and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 274) D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol.
  • inflammatory disorders immunologic disorders cancer Human interleukin-6 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R49041 WO9403492 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P05231/IL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-6 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 270) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16. inflammatory disorders immunologic disorders, cancer Mutant Interleukin 6 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, S176R GeneSeq monocytes, and macrophages.
  • Interleukin activity S176R variant of wildtype can be determined using assays known in the art: Matthews et al., in Lymphokines IL-6 which has the and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, sequence: D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
  • Interleukin-6 SEQ ID NO: 270
  • Interleukin 6 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R55256 monocytes, and macrophages.
  • Known functions include stimulating proliferation of JP06145063 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P05231/IL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-6 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 270) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16. inflammatory disorders immunologic disorders, cancer Interleukin 8 (IL-8) Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession R53932 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis JP06100595 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity GenBank: AAA59159.1 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 275) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80.
  • Soluble IL-8 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-7 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R59919 U.S. Pat. No. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis 5,328,988 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity P13232/IL7_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-7 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; and Park et al (1990) J. Exp. Med. 171, 1073-79. (SEQ ID NO: 276) inflammatory disorders immunologic disorders, cancer IL-3 containing fusion Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein. GeneSeq monocytes, and macrophages.
  • Interleukin activity Fusions of wildtype IL-3 can be determined using assays known in the art: Matthews et al., in Lymphokines which has the sequence: and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, P08700
  • Interleukin-3 inflammatory disorders immunologic disorders, cancer (SEQ ID NO: 449) IL-3 mutant proteins Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R79254, R79255, R79256, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis R79257, R79258, R79259, of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity R79260, R79261, R79262 can be determined using assays known in the art: Matthews et al., in Lymphokines R79263, R79264, R79265, and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, R79266, R79267, R79268, D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830.
  • Variants of wildtype IL-3 which has the sequence: P08700
  • Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Interleukin activity Interleukin-12 subunit beta can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 277) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders cancer AGF GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, R64240 WO9429344 monocytes, and macrophages.
  • Known functions include stimulating proliferation of Q8NI99/ANGL6_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Angiopoietin-related of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity protein 6 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 278) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders, cancer Human interlaukin-12 40 kD Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, subunit GeneSeq monocytes, and macrophages.
  • IL12B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-12 subunit beta and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 277) D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078.
  • Human interleukin-15 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor from clone P1 monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis R90843 WO9530695 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • I15RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-15 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit alpha D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830. Soluble IL-8 Isoform 1) receptor polypep- tides may be useful for inhibiting interleukin activities. Obesity. (SEQ ID NO: 453) Metabolic Disease. Diabetes.
  • Enhancing secretion and stability of Interleukin 15 Human interleukin-7 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R92796 WO9604306 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P13232/IL7_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-7 can be determined using assays known in the art: Matthews et al., in Lymphokines (isoform 1) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 276) D.C. 1987, pp. 221-225; and Park et al (1990) J. Exp. Med. 171, 1073-79.
  • inflammatory disorders immunologic disorders, cancer interleukin-9 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R92797 monocytes, and macrophages.
  • Interleukin activity Interleukin-9 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 279) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84.
  • cancer interleukin-3 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R92801 monocytes, and macrophages.
  • Known functions include stimulating proliferation of WO9604306 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P08700
  • Interleukin activity Interleukin-3 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 280) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
  • inflammatory disorders immunologic disorders, cancer Human interleukin-5 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Interleukin activity Interleukin-5 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 281) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
  • cancer Recombinant interleukin- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, 16 GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of W33373 DE19617202 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q14005/IL16_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Pro-interleukin-16 can be determined using assays known in the art: Matthews et al., in Lymphokines (isoform 1) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 282) D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70.
  • inflammatory disorders immunologic disorders, cancer Human IL-16 protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of W33234 DE19617202 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q14005/IL16_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Pro-interleukin-16 can be determined using assays known in the art: Matthews et al., in Lymphokines (isoform 1) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 282) D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol.
  • inflammatory disorders immunologic disorders
  • cancer Thrl 17 human interleukin Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, 9 GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of W27521 WO9708321 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P15248
  • Interleukin activity Interleukin-9 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 387) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders, cancer Metl 17 human interleukin Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, 9 GeneSeq Accession monocytes, and macrophages.
  • WO9708321 immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis SEQ ID NO: 388 of neutrophils and T lymphocytes
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84.
  • IL-1 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor antagonist. monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W77158 EP864585 (e.g. of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity SEQ ID NOs: 12 to 19, or can be determined using assays known in the art: Matthews et al., in Lymphokines 22 to 25 of this and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, publication). D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
  • inflammatory disorders immunologic disorders, cancer Human interleukin-18 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein (IL-18) GeneSeq monocytes, and macrophages.
  • Interleukin activity Q14116/IL18_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-18 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; and USHIO et al (1996) J. Immunol. 156, 4274-79.
  • inflammatory disorders immunologic disorders
  • cancer Human interleukin-18 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of W77077 EP861663 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q14116/IL18_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-18 can be determined using assays known in the art: Matthews et al., in Lymphokines (isoform 1) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 283) D.C. 1987, pp. 221-225; and USHIO et al (1996) J. Immunol. 156, 4274-79.
  • inflammatory disorders immunologic disorders, cancer Human interleukin 18 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, derivatives GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accessions W77083, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W77084, W77085, of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity W77086, W77087 can be determined using assays known in the art: Matthews et al., in Lymphokines W77088, and W77089 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, EP861663 D.C. 1987, pp. 221-225; and Ushio et al (1996) J.
  • IL18 inflammatory disorders immunologic disorders, cancer which is provided as: Q14116/IL18_HUMAN Interleukin-18 (isoform 1) (SEQ ID NO: 283) Interleukin-9 (IL-9) mature Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein (Thr117 version). monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W68158 WO9827997 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • FIG. 2 of WO9827997 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 389) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84.
  • inflammatory disorders immunologic disorders, cancer IL-9 mature protein variant Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, (Met117 version) GenSeq monocytes, and macrophages.
  • WO9827997 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 390) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84.
  • inflammatory disorders immunologic disorders cancer Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein variant #3. monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W64058 WO9824904 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Wildtype IL-9R is provided can be determined using assays known in the art: Matthews et al., in Lymphokines as: and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Q01113/IL9R_HUMAN D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84.
  • inflammatory Interleukin-9 receptor disorders immunologic disorders, cancer (isoform 1) (SEQ ID NO: 303)
  • Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein variant fragment monocytes, and macrophages.
  • Interleukin activity Wildtype IL-9R is provided can be determined using assays known in the art: Matthews et al., in Lymphokines as: and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Q01113/IL9R_HUMAN D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84.
  • Soluble IL-9 Interleukin-9 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • (isoform 1) (SEQ ID NO: 303)
  • Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein variant #3. monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W64061 WO9824904 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Wildtype IL-9R is provided can be determined using assays known in the art: Matthews et al., in Lymphokines as: and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Q01113/IL9R_HUMAN D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. Soluble IL-9 Interleukin-9 receptor receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human Interleukin-12 p40 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession W51311 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis WO9817689 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • IL12B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-12 subunit beta and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 277) D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078.
  • inflammatory disorders immunologic disorders, cancer Human Interleukin-12 p35 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein GeneSeq monocytes, and macrophages.
  • Interleukin activity P29459/IL12A_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-12 subunit and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, alpha D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078.
  • inflammatory disorders immunologic disorders, cancer Human protein with IL-16 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, activity GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession W63753 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis DE19649233- of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70.
  • inflammatory disorders immunologic disorders, cancer Human protein with IL-16 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, activity GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession W59425 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis DE19649233- of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70.
  • inflammatory disorders immunologic disorders cancer Human interleukin-15 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of W53878 U.S. Pat. No. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis 5,747,024 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity P40933/IL15_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-15 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform IL15-S48AA) D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830.
  • inflammatory SEQ ID NO: 285) disorders immunologic disorders, cancer. Obesity. Metabolic Disease. Diabetes.
  • Human wild-type Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, interleukin-4 (hIL-4) monocytes, and macrophages.
  • Known functions include stimulating proliferation of protein GeneSeq immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Accession W52149 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity WO9747744 can be determined using assays known in the art: Matthews et al., in Lymphokines P05112/IL4_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132, (isoform 1) 287-295.
  • inflammatory disorders immunologic disorders, cancer (SEQ ID NO: 286) interleukin-4 muteins Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accessions monocytes, and macrophages.
  • Known functions include stimulating proliferation of W52150, W52151, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W52153, W52154, of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity W52155, W52156 can be determined using assays known in the art: Matthews et al., in Lymphokines W52157, W52158, and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, W52159, W52160, D.C. 1987, pp.
  • WO9935268 immune cells
  • T helper cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis SEQ ID NO: 4 of of neutrophils and T lymphocytes e.g., chemotaxis SEQ ID NO: 4 of of neutrophils and T lymphocytes
  • interferons e.g., interferons.
  • Interleukin activity WO9935268 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 287) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
  • Human interleukin-1 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor antagonist beta monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Y24395 WO9935268 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
  • inflammatory disorders immunologic disorders, cancer Human EDIRF II protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, sequence GeneSeq monocytes, and macrophages.
  • Interleukin activity SEQ ID NO: 6 of can be determined using assays known in the art: Matthews et al., in Lymphokines WO9932632 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 391) D.C. 1987, pp. 221-225.
  • cancer Human EDIRF I protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, sequence GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession Y22197 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis WO9932632 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity SEQ ID NO: 2 of can be determined using assays known in the art: Matthews et al., in Lymphokines WO9932632 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 392) D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders, cancer Human IL- 1RD10 protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, sequence GeneSeq monocytes, and macrophages.
  • Interleukin activity SEQ ID NO: 20 of can be determined using assays known in the art: Matthews et al., in Lymphokines WO9919480 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
  • Soluble IL-1RD10 receptor polypep- tides may be useful for inhibiting interleukin activites.
  • Human IL- 1RD9 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of Y14122 WO9919480 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis SEQ ID NOS: 6, 8, 10 of of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity WO9919480 can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20. Soluble IL-1RD10 receptor polypep- tides may be useful for inhibiting interleukin activites.
  • Human DNAX interleukin- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, 40 GeneSeq Accession monocytes, and macrophages.
  • WO9919491 immune cells
  • immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis SEQ ID NO: 2 or 4 of of neutrophils and T lymphocytes and/or inhibition of interferons.
  • Interleukin activity WO9919491 can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 454) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders immunologic disorders
  • cancer cancer
  • Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, sequence GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession Y09197 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis WO9919491 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity SEQ ID NO: 4 of can be determined using assays known in the art: Matthews et al., in Lymphokines WO9919491 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 455) D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders, cancer IL-11 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, R50176 WO9405318 monocytes, and macrophages.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 288) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Lu et al (1994) J immunol. Methods 173, 19.
  • inflammatory disorders immunologic disorders cancer Human adipogenesis Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, inhibitory factor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession R43260 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis EP566410 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity also known as IL-11
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines P2080/
  • inflammatory disorders immunologic disorders, cancer isoform 1 (SEQ ID NO: 288)
  • IL-11 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, W02202 JP08127539 monocytes, and macrophages.
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 288) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Lu et al (1994) J immunol. Methods 173, 19.
  • cancer IL-14 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, R55800 WO9416074 monocytes, and macrophages.
  • Known functions include stimulating proliferation of P40222/TXLNA_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Alpha-taxilin of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity (SEQ ID NO: 289) can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Ambrus et al (1993) PNAS 90, 63330-34.
  • inflammatory disorders immunologic disorders, cancer IL-17 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession B03807 U.S. monocytes, and macrophages.
  • Known functions include stimulating proliferation of Pat. No.
  • Interleukin activity Interleukin-17 receptor A can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 290) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86.
  • Soluble IL- 17 receptor polypep- tides may be useful for inhibiting interleukin activites.
  • IL-17 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, R76573 WO9518826 monocytes, and macrophages.
  • Known functions include stimulating proliferation of Q16552/IL17_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Interleukin-17A of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity (SEQ ID NO: 291) can be determined using assays known in the art: Matthews et al., in Lymphokines and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86.
  • inflammatory disorders immunologic disorders, cancer CTLA-8 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession W13651 monocytes, and macrophages.
  • Interleukin activity Q16552/IL17_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-17A and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 291) D.C. 1987, pp. 221-225.
  • inflammatory disorders immunologic disorders cancer IL-19 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, W37935 WO9808870 monocytes, and macrophages.
  • Known functions include stimulating proliferation of Q9UHD0
  • Interleukin activity can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 292) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Gallagher et al (2000) Genes Immun. 1,442-50.
  • inflammatory disorders immunologic disorders, cancer IL-21 (TIF) GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession Y92879 monocytes, and macrophages.
  • Interleukin activity Interleukin-21 can be determined using assays known in the art: Matthews et al., in Lymphokines (isoform 1) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 293) D.C. 1987, pp.
  • inflammatory disorders immunologic disorders, cancer IL-8 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R33420 monocytes, and macrophages.
  • Known functions include stimulating proliferation of WO9306229 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis IL-8RA of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity P25024/CXCR1_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines C-X-C chemokine receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, type 1 D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8 (SEQ ID NO: 294) receptor polypep- tides may be useful for inhibiting interleukin activities.
  • IL-8RB P25025/CXCR2_HUMAN C-X-C chemokine receptor type 2 Human type II interleukin- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, 1 receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of Accession R85480 U.S. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Pat. No. 5,464,937 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • U.S. immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis Pat No. 5,464,937 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity P27930/IL1R2_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-1 receptor type 2 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 296) D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20. Soluble type II interleukin-1 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-12 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession R69632 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis EP638644 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity IL-12 receptor B1 can be determined using assays known in the art: Matthews et al., in Lymphokines P42701
  • Interleukin 8 receptor B Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of R80758 U.S. Pat. No. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis 5,440,021 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis 5,440,021 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity IL-8RB can be determined using assays known in the art: Matthews et al., in Lymphokines P25025/CXCR2_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, C-X-C chemokine receptor D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8 type 2 receptor B polypep- tides may be useful for inhibiting interleukin activities.
  • Human IL-8 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein hIL8RA GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession B09989 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis JP08103276 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity IL-8RA can be determined using assays known in the art: Matthews et al., in Lymphokines P25024/CXCR1_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, C-X-C chemokine receptor D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8 type 1 receptor A polypep- tides may be useful for inhibiting interleukin activities.
  • Human IL-8 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein hIL8R GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession B09990 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis JP08103276 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity IL-8RA can be determined using assays known in the art: Matthews et al., in Lymphokines P25024/CXCR1_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, C-X-C chemokine receptor D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8 type 1 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Interleukin-2 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, associated protein p43 monocytes, and macrophages. Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis R97569 WO9621732- of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity SEQ ID NO: 2 of can be determined using assays known in the art: Matthews et al., in Lymphokines WO9621732 and Interferons: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 395) D.C. 1987, pp. 221-225; and Gillis et al (1978) J. Immunol. 120, 2027. Soluble IL-2 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-17 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Interleukin activity Q96F46/I17RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-17 receptor A and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 290) D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol.
  • Soluble IL- 17 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-11 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession R99090 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis WO9619574 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Q14626/I11RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-11 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit alpha D.C. 1987, pp. 221-225; and Lu et al (1994) J immunol. Methods 173, 19. Soluble IL- (SEQ ID NO: 297) 11 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-1 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor accessory protein monocytes, and macrophages.
  • Known functions include stimulating proliferation of GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W01911 WO9623067 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Human IL1R Acp can be determined using assays known in the art: Matthews et al., in Lymphokines SEQ ID NO: 3 of and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, WO9623067 D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
  • AGF Protein GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R92749 U.S. monocytes, and macrophages.
  • Known functions include stimulating proliferation of Pat. No. 5,488,032 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q8NI99/ANGL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Angiopoietin-related can be determined using assays known in the art: Matthews et al., in Lymphokines protein 6 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 278) D.C. 1987, pp. 221-225.
  • Inflammatory disorders immunologic disorders, cancer Human interleukin-1 type- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, 3 receptor GeneSeq monocytes, and macrophages.
  • Interleukin activity SEQ ID NO: 2 and 4 of can be determined using assays known in the art: Matthews et al., in Lymphokines WO9607739 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 398 and SEQ D.C. 1987, pp.
  • IL-type-3 receptor polypep- tides may be useful for inhibiting interleukin activities
  • Human interleukin-13 beta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession W24972 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis WO9720926 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity SEQ ID NO: 2 from can be determined using assays known in the art: Matthews et al., in Lymphokines WO9720926 and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (SEQ ID NO: 400) D.C. 1987, pp. 221-225; and Boutelier et al (1995) J. Immunol. Methods 181, 29. Soluble IL-13 beta receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-13 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, alpha receptor GeneSeq monocytes, and macrophages.
  • Interleukin activity IL-13RA1 can be determined using assays known in the art: Matthews et al., in Lymphokines P78552/I13R1_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Interleukin-13 receptor D.C. 1987, pp. 221-225; and Boutelier et al (1995) J. Immunol.
  • Methods 181, 29. subunit alpha-1 Soluble IL-13 alpha receptor polypep- tides may be useful for inhibiting interleukin (isoform 1) activities.
  • IL-13RA2 Q14627/I13R2_HUMAN Interleukin-13 receptor subunit alpha-2 (SEQ ID NO: 299)
  • Human interleukin-4 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession W13499 U.S. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Pat. No.
  • Interleukin activity P24394/IL4RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-4 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit alpha D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132, (isoform 1) 287-295. Soluble IL-4 receptor polypep- tides may be useful for inhibiting interleukin (SEQ ID NO: 300) activities.
  • Human interleukin-12 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, beta-2 receptor GeneSeq monocytes, and macrophages.
  • Known functions include stimulating proliferation of Accession W12771 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis EP759466 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • I12R2_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-12 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit beta-2 D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. Soluble IL-12 (isoform 1) beta-2 receptor polypep- tides may be useful for inhibiting interleukin activities. (SEQ ID NO: 301) Human interleukin-12 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, beta-1 receptor.
  • GeneSeq monocytes, and macrophages are known functions.
  • Known functions include stimulating proliferation of Accession W12772 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis EP759466 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • I12R1_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-12 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit beta-1 D.C. 1987, pp.
  • Soluble IL-12 (isoform 1) beta-1 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Soluble IL-12 (isoform 1) beta-1 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • SEQ ID NO: 302 Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, protein GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of Accessions W64055, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis W64056, and W64057 of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis W64056, and W64057 of neutrophils and T lymphocytes and/or inhibition of inter
  • Interleukin activity WO9824904 can be determined using assays known in the art: Matthews et al., in Lymphokines Q01113/IL9R_HUMAN and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, Interleukin-9 receptor D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. Soluble IL-9 (isoform 1) receptor polypep- tides may be useful for inhibiting interleukin activities.
  • IL-10 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession W41804 U.S. monocytes, and macrophages.
  • Known functions include stimulating proliferation of Pat. No. 5,716,804 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis IL-10RA of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Q13651/I10R1_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-10 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit alpha D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510. (SEQ ID NO: 304) Soluble IL-10 receptor polypep- tides may be useful for inhibiting interleukin IL-10RB activities.
  • Human IL-6 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of Y30938 JP11196867 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P08887/IL6RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-6 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines subunit alpha and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16. (SEQ ID NO: 306) Soluble IL-6 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • II-17 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession Y97181 U.S.
  • Interleukin activity Interleukin-17 receptor A can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 290) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp.
  • Soluble IL- 17 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • II-17 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession Y97131 U.S. monocytes, and macrophages.
  • Known functions include stimulating proliferation of Pat. No. 6,100,235 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q96F46/I17RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-17 receptor A can be determined using assays known in the art: Matthews et al., in Lymphokines (SEQ ID NO: 290) and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86. Soluble IL- 17 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human interleukin-3 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, receptor GeneSeq monocytes, and macrophages.
  • Interleukin activity P26951/IL3RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-3 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit alpha D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
  • Soluble (isoform 1) IL-3 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • SEQ ID NO: 307 Human GM- CSF receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of R10919 WO9102063 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis GM-CSF receptor A of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • R10919 WO9102063 immune cells e.g., T helper cells, B cells, eosinophils, and lymphocytes
  • chemotaxis GM-CSF receptor A of neutrophils and T lymphocytes chemotaxis GM-CSF receptor A of neutrophils and T lymphocytes, and/or inhibition of interfer
  • Interleukin activity P15509/CSF2R_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Granulocyte-macrophage and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, colony-stimulating factor D.C. 1987, pp. 221-225. Soluble GM-CSF receptor polypep- tides may be useful for receptor subunit alpha inhibiting interleukin activities.
  • IL-5 receptor alpha Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, chain GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R25064 EP492214 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q01344/IL5RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-5 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines subunit alpha and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334. Soluble (SEQ ID NO: 310) IL-5 receptor alpha polypeptides may be useful for inhibiting interleukin activities.
  • II-5 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession W82842 monocytes, and macrophages.
  • Interleukin activity Interleukin-5 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines subunit alpha and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
  • Soluble (SEQ ID NO: 310) IL-5 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • II-6 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R37215 monocytes, and macrophages.
  • Known functions include stimulating proliferation of JP05091892 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P08887/IL6RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons.
  • Interleukin activity Interleukin-6 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines subunit alpha and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16. (SEQ ID NO: 306) Soluble IL-6 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • Human B cell stimulating Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, factor-2 receptor GeneSeq monocytes, and macrophages.
  • Interleukin activity P08887/IL6RA HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines Interleukin-6 receptor and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, subunit alpha D.C. 1987, pp. 221-225.
  • Soluble B cell stimulating factor-2 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • IL-7 receptor clone Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, GeneSeq Accession monocytes, and macrophages.
  • Known functions include stimulating proliferation of R08330 EP403114 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis P1687/
  • Interleukin activity Interleukin-7 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines subunit alpha and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; and Park et al (1990) J. Exp. Med. 171, 1073-79. Soluble IL- (SEQ ID NO: 311) 7 receptor polypep- tides may be useful for inhibiting interleukin activities.
  • EPO receptor; EPOR EPO Receptor is involved in the proliferation and differentiation of erythroblasts.
  • EPO GeneSeq Accession Receptor activity can be determined using assays known in the art, such as, J Biol R06512 WO9008822 Chem 2001 Mar. 23; 276(12: 8995-9002; JAK2 protein tyrosine kinase activity: Blood P19235/EPOR_HUMAN 1994 Sep. 1; 84(5): 1501-7 and Mol Cell Biol. 1994 October; 14(10: 6506-14.
  • Inflammatory Erythropoietin receptor disorders immunologic disorders, cancer, erythroblast proliferation and differentiation (isoform EPOR-F) (SEQ ID NO: 312)
  • IL-15 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes, Accession R90843 monocytes, and macrophages.
  • Known functions include stimulating proliferation of WO9530695 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis Q1326/
  • Interleukin activity Interleukin-15 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines subunit alpha and Interferens: A Practical Approach , Clemens et al., eds, IRL Press, Washington, (isoform 1) D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830. Soluble IL-15 (SEQ ID NO: 313) receptor polypep- tides may be useful for inhibiting interleukin activities. Obesity. Metabolic Disease. Diabetes.
  • Apoptosis activity, NF-kB activation, and B and T cell co- Accession R70977 stimulation can be determined using assays known in the art: Moore et al., 1999, WO9507984 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Q07011/TNR9_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Soluble 4-1BB Tumor necrosis factor receptor polypeptides may be useful for inhibiting apoptosis, NF-kB activation, receptor superfamily and/or co-stimulation of immune cells such as B and T cells.
  • member 9 SEQ ID NO: 314) BCMA GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession Y71979 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO0068378 stimulation can be determined using assays known in the art: Moore et al., 1999, Q02223/TNR17_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble BCMA receptor superfamily receptor polypeptides may be useful for inhibiting apoptosis, NF-kB activation, member 17 and/or co-stimulation of immune cells such as B and T cells.
  • isoform 1 (SEQ ID NO: 315) CD27 GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune R20814 WO9201049 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- P26842/CD27_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, CD27 antigen Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): (SEQ ID NO: 316) 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol.
  • Soluble CD27 polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co- stimulation of immune cells such as B and T cells.
  • CD30 GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune R35478 DE4200043 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- P28908/TNR8_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble CD30 member 8 polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co- (isoform 1) stimulation of immune cells such as B and T cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- P25942/TNR5_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Soluble CD40 member 5 polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co- (isoform 1) stimulation of immune cells such as B and T cells.
  • SEQ ID NO: 318 EDAR Genbank Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession AAD50077 cells such as T and B cells.
  • EDAR_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Immune Disorders member EDAR Lymphomas, X-linked hypohidrotic ectodermal dysplasia (isoform 1) (SEQ ID NO: 319) OX40; ACT-4 GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession R74737 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9512673 stimulation can be determined using assays known in the art: Moore et al., 1999, P43489/TNR4_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Immune Disorders TACI GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune W75783 WO9839361 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- O14836/TR13B_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble TACI member 13B receptor polypep- tides may be useful for inhibiting apoptosis, NF-kB activation, (isoform 1) and/or co-stimulation of immune cells such as B and T cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- AU9058976 stimulation can be determined using assays known in the art: Moore et al., 1999, P19438/TNR1A_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Soluble TNF-R receptor superfamily receptor polypep- tides may be useful for inhibiting apoptosis, NF-kB activation, member 1A and/or co-stimulation of immune cells such as B and T cells.
  • (isoform 1) SEQ ID NO: 322) TNF-RII; TNF p75 Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune receptor; Death Receptor cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- GeneSeq Accession stimulation can be determined using assays known in the art: Moore et al., 1999, R11141 EP418014 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): P20333/TNR1B_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Soluble TNFR-II Tumor necrosis factor receptor polypep- tides may be useful for inhibiting apoptosis, NF-kB activation, receptor superfamily and/or co-stimulation of immune cells such as B and T cells.
  • member 1B isoform 1 (SEQ ID NO: 323) hAPO-4; TROY GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession W93581 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9911791 stimulation can be determined using assays known in the art: Moore et al., 1999, Q9N568/TNR19_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Immune Disorders receptor superfamily Cancers member 19 (isoform 1) (SEQ ID NO: 324) TNF-alpha precursor Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune GeneSeq Accession cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- P60074 EP205038 stimulation can be determined using assays known in the art: Moore et al., 1999, Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory disorders immunologic disorders, cancer Human TNF- alpha Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune GeneSeq Accession cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- R62463 EP619372 stimulation can be determined using assays known in the art: Moore et al., 1999, P01375/TNFA_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory SEQ ID NO: 325) disorders immunologic disorders, cancer Human TNF- alpha Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune GeneSeq Accession cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- R42679 EP563714 stimulation can be determined using assays known in the art: Moore et al., 1999, P01375/TNFA_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol.
  • Inflammatory SEQ ID NO: 325) disorders immunologic disorders, cancer Human TNF- beta (LT- Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune alpha) GeneSeq cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- Accession B37799 stimulation can be determined using assays known in the art: Moore et al., 1999, WO0064479 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): P01374/TNFB_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory Lymphotoxin-alpha disorders immunologic disorders, cancer (SEQ ID NO: 326) LT-alpha GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession P70107 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- EP250000 stimulation can be determined using assays known in the art: Moore et al., 1999, P01374/TNFB_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Lymphotoxin-alpha 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory SEQ ID NO: 326 disorders immunologic disorders, cancer LT-beta GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession R56869 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9413808 stimulation can be determined using assays known in the art: Moore et al., 1999, Q06643/TNFC_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Lymphotoxin-beta 9792-6; Epsevik and Nissen-Meyer, 1986, J.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9846751 stimulation can be determined using assays known in the art: Moore et al., 1999, O14788/TNF11_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory ligand superfamily disorders immunologic disorders, cancer, loss of bone mass member 11 (isoform 1) (SEQ ID NO: 328) FasL GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune W98071 WO9903999 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- P48023/TNFL6_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): ligand superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory member 6 disorders immunologic disorders, cancer (isoform 1) (SEQ ID NO: 329) FasL GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune W95041 WO9903998 cells such as T and B cells.
  • cancer isoform 1 (SEQ ID NO: 329) FasL GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune W95041 WO9903998 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- P48023/TNFL6_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999, Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): ligand superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory member 6 disorders immunologic disorders, cancer (isoform 1) (SEQ ID NO: 329) CD27L GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession R50121 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9405691 stimulation can be determined using assays known in the art: Moore et al., 1999, P32970/CD70_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): CD70 antigen 9792-6; Epsevik and Nissen-Meyer, 1986, J.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9324135 stimulation can be determined using assays known in the art: Moore et al., 1999, P32971/TNFL8_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory ligand superfamily disorders immunologic disorders, cancer member 8 (SEQ ID NO: 331) CD40L GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Accession R85486 cells such as T and B cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9529935 stimulation can be determined using assays known in the art: Moore et al., 1999, P29965/CD40L_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): CD40 ligand 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • Inflammatory SEQ ID NO: 332
  • cancer 4-1BB ligand GeneSeq Activities associated with apoptosis NF-kB activation
  • co- stimulation of immune Accession W26657 U.S. cells such as T and B cells.
  • 5,674,704 stimulation can be determined using assays known in the art: Moore et al., 1999, P41273/TNFL9_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory ligand superfamily disorders immunologic disorders, cancer member 9 (SEQ ID NO: 333) FAS Ligand Inhibitory Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune Protein (DcR3) GeneSeq cells such as T and B cells.
  • DcR3 immune Protein
  • Apoptosis activity, NF-kB activation, and B and T cell co- Accession B19335 stimulation can be determined using assays known in the art: Moore et al., 1999, WO0058465 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): O95407/TNF6B_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble DcR3 Tumor necrosis factor polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co- receptor superfamily stimulation of immune cells such as B and T cells.
  • Apoptosis activity, NF-kB activation, and B and T cell co- WO9521915 stimulation can be determined using assays known in the art: Moore et al., 1999, P23510/TNFL4_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18): Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods.
  • HIV protease assays EP0387231.
  • Retroviral protease Peptides that inhibit the function/binding of HIV HIV protease activities are known in inhibitors GeneSeq the art: HIV protease assays: EP0387231. One can modify the assay to look for Accessions R06660, inhibition using any of the disclosed protease inhibitor polypeptides.
  • HIV HIV, R86329, R86330, R86331, inflammatory disorders, immuno- logic disorders, cancer, viral infections R86332, R86333, R86334, R86335, R86336, R86337, R86338, R86339, R86340, R86341, R86342, R86343, R86344, R86345, R86346, R86347, R86348, R86349, R86350, R86351, R86352, R86353, R86354, R86355, R86356, R86357, R86358, R86359, R86360, R86361, R86362, R86363, R86364, R86365, R86366, R86367, R86368, R86369, R86370, and R86371 DE4412174 HIV Inhibitor Peptide Peptides that inhibit the function/
  • HIV inflammatory disorders, immuno- logic disorders, cancer, viral infections HIV Inhibitor Peptide Peptides that inhibit the function/binding of HIV HIV protease activities are known in GenSeq Accession the art: HIV protease assays: EP0387231.
  • HIV protease assays EP0387231.
  • HIV, inflammatory disorders, immuno- logic disorders, cancer, viral infections HIV Inhibitor Peptide Peptides that inhibit the function/binding of HIV HIV protease activities are known in Science 291, 884 (2001); the art: HIV protease assays: EP0387231.
  • HIV, 2001; 10.1126/science.1057453 inflammatory disorders, immuno- logic disorders, cancer, viral infections
  • Human monocyte Chemokines are a family of related small, secreted proteins involved in biological chemoattractant factor processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • hMCP-3 GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession R73915 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9509232 chemokines exert their effects by acting on a family of seven transmembrane G- P80098/CCL7_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-C motif chemokine 7 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 336) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C.
  • Human monocyte Chemokines are a family of related small, secreted proteins involved in biological chemoattractant factor processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • hMCP-1 GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession R73914 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9509232 chemokines exert their effects by acting on a family of seven transmembrane G- P13500/CCL2_HUMAN protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 337) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accessions R66699 and Members of this family are involved in a similarly diverse range of pathologies W17671 WO9429341 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P19875/CXCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-X-C motif chemokine 2 protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Accessions R66700 and Members of this family are involved in a similarly diverse range of pathologies W17672 WO9429341 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P19876/CXCL3_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-X-C motif chemokine 3 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 339) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accessions R66698 and Members of this family are involved in a similarly diverse range of pathologies W18024 WO9429341 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P09341/GROA_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- Growth-regulated alpha protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 340) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, inflammatory disorders, blood- related disorders, stem cell transplantation, cancer Human eosinophil- Chemokines are a family of related small, secreted proteins involved in biological expressed chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • EEC EEC GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession W05186 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9632481 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 2 of protein coupled receptors. Over 40 human chemokines have been described, which WO9632481 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 401) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A.
  • Chemokines are a family of related small, secreted proteins involved in biological PF4-414 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions R92318 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the R99809 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G- FIG.
  • Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological 8M3 GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. R99812 WO9613587 Members of this family are involved in a similarly diverse range of pathologies including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80. Cancer and blood- related disorders, particularly myelosuppression Human interleukin-8 (IL-8) Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • IL-8 Human interleukin-8
  • R99814 WO9613587 Members of this family are involved in a similarly diverse range of pathologies P10145/IL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Interleukin-8 chemokines exert their effects by acting on a family of seven transmembrane G- (isoform 1) protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 341) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological 8M1 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions R99815 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the R99803 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G- FIG.
  • Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological 8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions R99816 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the R99805 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G- FIG. 4C of WO9613587 protein coupled receptors.
  • chemokines Over 40 human chemokines have been described, which (SEQ ID NO: 404) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
  • Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological 8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions R99817 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the R99806 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G- FIG. 4C of WO9613587 protein coupled receptors.
  • chemokines Over 40 human chemokines have been described, which (SEQ ID NO: 404) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80. Cancer and blood- related disorders, particularly myelosuppression.
  • Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological 8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions R99818 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the R99804 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G- FIG. 4C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 404) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80. Cancer and blood- related disorders, particularly myelosuppression.
  • Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological 8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological 8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions R99822 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the R9807 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G- FIG. 4C of WO9613587 protein coupled receptors.
  • Chemokines Over 40 human chemokines have been described, which (SEQ ID NO: 404) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer and blood- related disorders, particularly myelosuppression. Human foetal spleen expressed Chemokines are a family of related small, secreted proteins involved in biological chemo-kine, processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • FSEC GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession R98499 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9622374 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 2 of protein coupled receptors. Over 40 human chemokines have been described, which WO9622374 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 405) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A.
  • Liver expressed Chemokines are a family of related small, secreted proteins involved in biological chemokine-1(LVEC-1) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies R95689 WO9616979 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9616979 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 406) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Inflammation of the liver Liver expressed Chemokines are a family of related small, secreted proteins involved in biological chemokine-2(LVEC-2) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies R95690 WO9616979 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 4 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9616979 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 407) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine (PGEC) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies R95691 WO9616979 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 6 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9616979 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 408) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Inflammation, particularly of the liver Adenoid-expressed Chemokines are a family of related small, secreted proteins involved in biological chemokine (ADEC) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • ADEC biological chemokine
  • chemokine CC-2 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W38170 WO9741230 Members of this family are involved in a similarly diverse range of pathologies Q16663/CCL15_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 15 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 342) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, cell migration, proliferation, and differentiation disorders Human chemokine HCC-1 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W38171 WO9741230 Members of this family are involved in a similarly diverse range of pathologies Q16627/CCL14_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 14 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 343) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • chemokine CC- 3 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W38172 WO9741230 Members of this family are involved in a similarly diverse range of pathologies Q16627/CCL14_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 14 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 343) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, cell migration, proliferation, and differentiation Novel betachemokine Chemokines are a family of related small, secreted proteins involved in biological designated PTEC processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9739126 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 410) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological acid chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W23344 Members of this family are involved in a similarly diverse range of pathologies WO9727299 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9727299 protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W25942 WO9721812 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 4 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9721812 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 412) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Human beta- chemokine Chemokines are a family of related small, secreted proteins involved in biological H1305 (MCP-2) GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W26655 Members of this family are involved in a similarly diverse range of pathologies WO9725427 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P80075/CCL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 8 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemotaxis blood- related disorders, viral infection, HIV, wound healing, cancer Human eosinocyte CC Chemokines are a family of related small, secreted proteins involved in biological type chemokine eotaxin processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W14990 WO9712914 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The P51671/CCL11_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- Eotaxin protein coupled receptors.
  • chemokines Over 40 human chemokines have been described, which (SEQ ID NO: 245) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Inflammatory and immune disorders Human thymus and Chemokines are a family of related small, secreted proteins involved in biological activation regulated processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • cytokine (TARC) GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession W14018 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9711969 chemokines exert their effects by acting on a family of seven transmembrane G- Q92583/CCL17_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-C motif chemokine 17 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 261) using assays known in the art: Methods in molecular Biology, 2000, vol. 138: Chemokine Protocols, Edited by A. E. I. Proudfoot, T. N. C.
  • Human chemokine beta-8 Chemokines are a family of related small, secreted proteins involved in biological short forms GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W16315 Members of this family are involved in a similarly diverse range of pathologies WO9712041 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Wildtype chemokine beta- chemokines exert their effects by acting on a family of seven transmembrane G- 8 provided as: protein coupled receptors.
  • Chemokine activities can be determined C-C motif chemokine 23 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: (SEQ ID NO: 459) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Microphage derived Chemokines are a family of related small, secreted proteins involved in biological chemokine, MDC processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W20058 WO9640923 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the O00626/CCL22_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 22 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 345) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Inflammatory diseases, wound healin, angiogenesis Human chemokine ZSIG- Chemokines are a family of related small, secreted proteins involved in biological 35 GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W30565 WO9844117 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 2 of WO including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9844117 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 413) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological “ILINCK” GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accesssion W69990 Chemokine activities can be determined using assays known in the art: Methods in WO98328658 Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. SEQ ID NO: 4 from Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • WO9832858 Immune and inflammatory disorders, abnormal proliferation, regen- eration, (SEQ ID NO: 414) generation and atrophy disorders
  • SEQ ID NO: 4114 Primate CXC chemokine Chemokines are a family of related small, secreted proteins involved in biological “IBICK” GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W69989 Members of this family are involved in a similarly diverse range of pathologies WO9832858 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 2 from chemokines exert their effects by acting on a family of seven transmembrane G- WO9832858 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders, abnormal proliferation, regen- eration, generation and atrophy disorders Human CC-type Chemokines are a family of related small, secreted proteins involved in biological chemokine protein processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • SLC Members of this family are involved in a similarly diverse range of pathologies (secondary lymphoid including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the chemokine) GeneSeq chemokines exert their effects by acting on a family of seven transmembrane G- Accession W69163 protein coupled receptors. Over 40 human chemokines have been described, which WO9831809 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined O00585/CCL21_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: C-C motif chemokine 21 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N.
  • Chemokines are a family of related small, secreted proteins involved in biological ELC protein GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W62542 Members of this family are involved in a similarly diverse range of pathologies WO9826071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Q99731/CCL19_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 19 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer and infectious diseases, particularly herpes virus Human DVic-1 C-C Chemokines are a family of related small, secreted proteins involved in biological chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9823750 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 416) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological DGWCC GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W60650 Members of this family are involved in a similarly diverse range of pathologies WO9823750 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 6 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9823750 protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological Accession W62783 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9824907 Members of this family are involved in a similarly diverse range of pathologies O00626/CCL22_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 22 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 345) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Immune disorders particularly T cell related disorders, viral infection, and inflammation, especially joint Exodus protein GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W61279 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9821330 Members of this family are involved in a similarly diverse range of pathologies P78556/CCL20_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 20 chemokines exert their effects by acting on a family of seven transmembrane G- (isoform 1) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • WO9814581 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 10 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9814581 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 418) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer and de- generative disorders Human T cell mixed Chemokines are a family of related small, secreted proteins involved in biological lymphocyte reaction processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. expressed chemokine Members of this family are involved in a similarly diverse range of pathologies (TMEC) GeneSeq including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • TMEC pathologies
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W50885 W09814581 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 8 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9814581 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 419) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer and de- generative disorders human liver and activation
  • Chemokines are a family of related small, secreted proteins involved in biological regulated chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. (LARC) GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession W57475 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W29538 WO9744462 Members of this family are involved in a similarly diverse range of pathologies Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the herien as chemokines exert their effects by acting on a family of seven transmembrane G- P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-C motif chemokine 5 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Infectious diseases, particularly HIV RANTES 8-68 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W29529 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9744462 Members of this family are involved in a similarly diverse range of pathologies Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the herein as chemokines exert their effects by acting on a family of seven transmembrane G- P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-C motif chemokine 5 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A.
  • Infectious diseases, particularly HIV RANTES 9-68 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession W29528 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. WO9744462 Members of this family are involved in a similarly diverse range of pathologies Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the herien as chemokines exert their effects by acting on a family of seven transmembrane G- P13501/CCL5_HUMAN protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Infectious diseases, particularly HIV Human chemokine protein Chemokines are a family of related small, secreted proteins involved in biological 331D5 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • SEQ ID NO: 12 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9811226 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 420) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological 61164 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W59430 Members of this family are involved in a similarly diverse range of pathologies WO9811226 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 6 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9811226 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Abnormal prolifera- tion, regeneration, degeneration, or atrophy, including cancer Chemokine MCP-4 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W56690 WO9809171 Members of this family are involved in a similarly diverse range of pathologies Q99616/CCL13_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 13 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 347) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Human stromal cell- Chemokines are a family of related small, secreted proteins involved in biological derived chemokine, SDF-1 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W50766 FR2751658 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P48061/SDF1_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- Stromal cell-derived factor 1 protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 260) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. HIV infections Thymus expressed Chemokines are a family of related small, secreted proteins involved in biological chemokine (TECK) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • TECK biological chemokine
  • Human chemokine MIP- Chemokines are a family of related small, secreted proteins involved in biological 3alpha GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W44398 Members of this family are involved in a similarly diverse range of pathologies WO9801557 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P78556/CCL20_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 20 protein coupled receptors.
  • chemokines Over 40 human chemokines have been described, which (isoform 1) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined (SEQ ID NO: 248) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders Human chemokine MIP- Chemokines are a family of related small, secreted proteins involved in biological 3beta GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W44399 WO9801557 Members of this family are involved in a similarly diverse range of pathologies Q99731/CCL19_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 19 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 249) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Human monocyte Chemokines are a family of related small, secreted proteins involved in biological chemotactic proprotein processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. (MCPP) sequence Members of this family are involved in a similarly diverse range of pathologies GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the W42072 WO9802459 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 1 of protein coupled receptors. Over 40 human chemokines have been described, which WO9802459 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 456) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Immune disorders, respiratory disorders, cancer Macrophage- derived Chemokines are a family of related small, secreted proteins involved in biological chemokine (MDC) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accessions Members of this family are involved in a similarly diverse range of pathologies W40811 and Y24414 U.S.
  • cancer Macrophage derived Chemokines are a family of related small, secreted proteins involved in biological chemokine analogue processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. MDC-eyfy GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession Y24416 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Pat. No. 5,932,703 (“eyfy” chemokines exert their effects by acting on a family of seven transmembrane G- disclosed as SEQ ID NO: protein coupled receptors.
  • Macrophage derived Chemokines are a family of related small, secreted proteins involved in biological chemokine analogue MDC processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. (n + 1) GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession Y24413 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Pat. No. 5,932,703 chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
  • Macrophage derived Chemokines are a family of related small, secreted proteins involved in biological chemokine analogue processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. MDC-yl GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession Y24415 U.S.
  • chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine eotaxin 3 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. protein sequence Members of this family are involved in a similarly diverse range of pathologies GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Y43178 JP11243960 chemokines exert their effects by acting on a family of seven transmembrane G- Q9Y258/CCL26_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-C motif chemokine 26 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 349) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Allergic diseases and HIV infection Human MCP-3 and human Chemokines are a family of related small, secreted proteins involved in biological Muc-1 core epitope (VNT) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • VNT biological Muc-1 core epitope
  • fusion protein GeneSeq Members of this family are involved in a similarly diverse range of pathologies Acession Y29893 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9946392 chemokines exert their effects by acting on a family of seven transmembrane G- Wildtype MCP-3 has the protein coupled receptors. Over 40 human chemokines have been described, which sequence: bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined P80098/CCL7_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: C-C motif chemokine 7 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C.
  • the WO9946392 chemokines exert their effects by acting on a family of seven transmembrane G- Wildtype IP10 has the protein coupled receptors. Over 40 human chemokines have been described, which sequence: bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined P02778/CXL10_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: C-X-C motif chemokine 10 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. (SEQ ID NO: 242) Humana Press Inc., Totowa, NJ.
  • HIV Wildtype Muc-1 has the infection sequence: P15941
  • Chemokines are a family of related small, secreted proteins involved in biological gp 120 hyper- variable processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. region fusion protein Members of this family are involved in a similarly diverse range of pathologies GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Y29897 W09946392 chemokines exert their effects by acting on a family of seven transmembrane G- Wildtype IP10 has the protein coupled receptors.
  • Chemokine activities can be determined P02778/CXL10_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: C-X-C motif chemokine 10 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. (SEQ ID NO: 242) Humana Press Inc., Totowa, NJ.
  • HIV Wildtype gp120 has the infection sequence: P03378
  • Human mammary Chemokines are a family of related small, secreted proteins involved in biological associated chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. (MACK) protein Full- Members of this family are involved in a similarly diverse range of pathologies Length and Mature including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the GeneSeq Accessions chemokines exert their effects by acting on a family of seven transmembrane G- Y29092 and Y29093 protein coupled receptors.
  • Chemokine activities can be determined Full-length: SEQ ID NO: 1 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: of WO9936540 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. (SEQ ID NO: 423) Humana Press Inc., Totowa, NJ.
  • Tim-1 protein GeneSeq Chemokines are a family of related small, secreted proteins involved in biological Accession Y28290 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9933990 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9933990 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 350) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Inflammation due to stimuli such as heart attacks and stroke, infection, physical trauma, UV or ionizing radiation, burns, frostbite or corrosive chemicals Human Lkn-1 Full-Length Chemokines are a family of related small, secreted proteins involved in biological and Mature protein processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine met- hSDF-1 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. alpha GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y05818 WO9920759 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 10 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9920759 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 425) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine met- hSDF-1 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. beta GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y05819 WO9920759 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 11 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9920759 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 426) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Inhibit or stimulate angiogenesis, inhibit the binding of HIV, antiinflammatory; immunosuppressant N-terminal modified Chemokines are a family of related small, secreted proteins involved in biological chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • GroHEK/hSDF- 1alpha Members of this family are involved in a similarly diverse range of pathologies GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Y05820 WO9920759 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 12 of protein coupled receptors. Over 40 human chemokines have been described, which WO9920759 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 427) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C.
  • immunosuppressant N-terminal modified Chemokines are a family of related small, secreted proteins involved in biological chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GroHEK/hSDF- 1beta. Members of this family are involved in a similarly diverse range of pathologies GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Y05821 WO9920759 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 13 of protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 428) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • immunosuppressant Chemokine Eotaxin Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Y14230 WO9912968 Members of this family are involved in a similarly diverse range of pathologies P51671/CCL11_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Eotaxin chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 245) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokine hMCP1a Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Y14225 WO9912968 Members of this family are involved in a similarly diverse range of pathologies including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound healing, cancer, prevent organ transplant rejection, Increase or enhance an inflammatory response, Chemokine hMCP1b Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Y14226 WO9912968 Members of this family are involved in a similarly diverse range of pathologies including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokine hSDF1b Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Y14228 WO9912968 Members of this family are involved in a similarly diverse range of pathologies P48061/SDF1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Stromal cell-derived factor 1 chemokines exert their effects by acting on a family of seven transmembrane G- (isoform beta) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokine hIL-8 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Y14229 WO9912968 Members of this family are involved in a similarly diverse range of pathologies P10145/IL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Interleukin-8 chemokines exert their effects by acting on a family of seven transmembrane G- (isoform 1) protein coupled receptors.
  • chemokines Over 40 human chemokines have been described, which (SEQ ID NO: 341) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
  • Chemokine hMCP1 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Y14222 WO9912968 Members of this family are involved in a similarly diverse range of pathologies P13500/CCL2_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 2 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 337) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound healing, cancer, prevent organ transplant rejection, Increase or enhance an inflammatory response, Chemokine hMCP2 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Y14223 WO9912968 Members of this family are involved in a similarly diverse range of pathologies P80075/CCL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 8 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 344) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokine hMCP3 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Y14224 WO9912968 Members of this family are involved in a similarly diverse range of pathologies P80098/CCL7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 7 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 336) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound healing, cancer, prevent organ transplant rejection, Increase or enhance an inflammatory response, C-C chemokine, MCP2 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Wild type monocyte Chemokines are a family of related small, secreted proteins involved in biological chemotactic protein 2 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y07233 EP906954 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P80075/CCL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 8 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 344) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Truncated monocyte Chemokines are a family of related small, secreted proteins involved in biological chemotactic protein 2 (6- processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. 76) GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y07234 EP906954 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the FIG. 1 of EP905241 and chemokines exert their effects by acting on a family of seven transmembrane G- EP906954 protein coupled receptors.
  • chemokines Over 40 human chemokines have been described, which (SEQ ID NO: 429) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A.
  • Truncated RANTES Chemokines are a family of related small, secreted proteins involved in biological protein (3-68) GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accessions Y07236 and Members of this family are involved in a similarly diverse range of pathologies Y07232 EP905241; including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the EP906954 chemokines exert their effects by acting on a family of seven transmembrane G- FIG.
  • Wild type monocyte Chemokines are a family of related small, secreted proteins involved in biological chemotactic protein 2 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y07237 EP905241 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • Truncated monocyte Chemokines are a family of related small, secreted proteins involved in biological chemotactic protein 2 (6-76) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y07238 EP905241 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • FIG. 1 The FIG.
  • a partial CXCR4B protein Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • W97363 EP897980 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the EP897980 chemokines exert their effects by acting on a family of seven transmembrane G- (sEQ ID NO: 431) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power, Humana Press Inc., Totowa, NJ Soluble CXCR4B receptor polypep- tides may be useful for inhibiting chemokine activities and viral infection.
  • Interferon gamma- Chemokines are a family of related small, secreted proteins involved in biological inducible protein (IP-10) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W96709 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- P02778/CXL10_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-X-C motif chemokine 10 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 242) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power, Humana Press Inc., Totowa, NJ Angiogenesis, Cancer, Inflammatory and Immune disorders, Cardio- Vascular disorders, Musco-skeletal disorders
  • a monokine induced by Chemokines are a family of related small, secreted proteins involved in biological gamma- interferon (MIG) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • MIG biological gamma- interferon
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W96711 U.S. Pat. No. Members of this family are involved in a similarly diverse range of pathologies 5,871,723 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P10145/IL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- Interleukin-8 protein coupled receptors. Over 40 human chemokines have been described, which (isoform 1) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined (SEQ ID NO: 341) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
  • Epithelial neutrophil Chemokines are a family of related small, secreted proteins involved in biological activating protein-78 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. (ENA-78) GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accession W96712 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Pat. No. 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- P42830/CXCL5_HUMAN protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 352) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power, Humana Press Inc., Totowa, NJ Angiogenesis, Cancer, Inflammatory and Immune disorders, Cardio- Vascular disorders, Musco-skeletal disorders Growth related oncogene- Chemokines are a family of related small, secreted proteins involved in biological alpha (GRO-alpha).
  • oncogene- Chemokines are a family of related small, secreted proteins involved in biological beta (GRO-beta). processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W96714 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- P19875/CXCL2_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which C-X-C motif chemokine 2 bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined (SEQ ID NO: 338) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • GRO-gamma biological gamma
  • angiogenesis a family of related small, secreted proteins involved in biological gamma (GRO-gamma) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • GRO-gamma biological gamma
  • GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W96715 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- P19876/CXCL3_HUMAN protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 339) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • a platelet basic protein Chemokines are a family of related small, secreted proteins involved in biological (PBP) GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W96716 U.S. Members of this family are involved in a similarly diverse range of pathologies Pat. No. 5,871,723 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • PBP protein-bindse
  • the P02775/CXCL7_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- Platelet basic protein protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological activating protein-III processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • CRISPR-III GeneSeqAccession Members of this family are involved in a similarly diverse range of pathologies S96717 U.S. Pat. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the No. 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 9 of U.S. protein coupled receptors. Over 40 human chemokines have been described, which Pat. No. 5,871,723 bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined (SEQ ID NO: 354) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I.
  • Chemokines are a family of related small, secreted proteins involved in biological protein (beta-TG) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W96718 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 10 of U.S. protein coupled receptors. Over 40 human chemokines have been described, which Pat. No. 5,871,723 bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined (SEQ ID NO: 355) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological peptide-2 (NAP-2) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W96719 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G- SEQ ID NO: 11 of U.S. protein coupled receptors. Over 40 human chemokines have been described, which Pat. No.
  • Chemokine activities can be determined (SEQ ID NO: 356) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Granulocyte chemotactic Chemokines are a family of related small, secreted proteins involved in biological protein-2 (GCP-2) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • GCP-2 biological protein-2
  • chemokine MIG- Chemokines are a family of related small, secreted proteins involved in biological beta protein GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession W90124 Members of this family are involved in a similarly diverse range of pathologies EP887409 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the (SEQ ID NO: 463) chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, viral, parasitic, fungal or bacterial infections, Cancer; autoimmune diseases or transplant rejection Human ZCHEMO-8 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9854326 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 432) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W82717 WO9854326 Members of this family are involved in a similarly diverse range of pathologies P13236/CCL4_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 4 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 358) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Acession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W82720 WO9854326 Members of this family are involved in a similarly diverse range of pathologies P13501/CCL5_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 5 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 241) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W82721 WO9854326 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 37 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 433) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W82722 W09854326 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 38 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 434) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. W82723 WO9854326 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 39 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 435) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders, autoimmune disorders and immunodeficiencies, Inflammatory and infectious diseases, Vascular disorders, wound healing A human L105 chemokine Chemokines are a family of related small, secreted proteins involved in biological designated huL105_3.
  • chemokines exert their effects by acting on a family of seven transmembrane G- WO9856818 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 436) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I.
  • a human L105 chemokine Chemokines are a family of related small, secreted proteins involved in biological designated huL105_7. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies W87589 WO9856818 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 4 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9856818 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer, wound healing Human mature gro-alpha Chemokines are a family of related small, secreted proteins involved in biological polypeptide used to treat processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • WO9848828 chemokines exert their effects by acting on a family of seven transmembrane G- P19876/CXCL3_HUMAN protein coupled receptors.
  • Chemokine activities can be determined (SEQ ID NO: 339) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Infectious diseases, sepsis Human thymus expressed Chemokines are a family of related small, secreted proteins involved in biological chemokine TECK and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • TECK variant GeneSeq Members of this family are involved in a similarly diverse range of pathologies Accessions B19607 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the B19608 WO0053635 chemokines exert their effects by acting on a family of seven transmembrane G- Wildtype TECK provided protein coupled receptors. Over 40 human chemokines have been described, which as: bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined O15444/CCL25_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: C-C motif chemokine 25 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N.
  • chemokine Chemokines are a family of related small, secreted proteins involved in biological SDF1alpha GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession B15791 Members of this family are involved in a similarly diverse range of pathologies WO0042071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P48061-2/SDF1_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- Isoform Alpha of Stromal protein coupled receptors.
  • Chemokine activities can be determined (isoform alpha) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: (SEQ ID NO: 259) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Human chemokine GRO- Chemokines are a family of related small, secreted proteins involved in biological alpha GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15793 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P09341/GROA_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Growth-regulated alpha chemokines exert their effects by acting on a family of seven transmembrane G- protein protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 340) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and Inflammatory disorders Human chemokine eotaxin Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • B15794 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P51671/CCL11_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Eotaxin chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 245) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Human chemokine MIG Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15803 WO0042071 Members of this family are involved in a similarly diverse range of pathologies Q07325/CXCL9_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-X-C motif chemokine 9 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 351) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and Inflammatory disorders Human chemokine PF4 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • B15804 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P02776/PLF4_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Platelet factor 4 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 359) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15805 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P22362/CCL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-C motif chemokine 1 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 360) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and Inflammatory disorders Human chemokine HCC-1 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • C-C motif chemokine 14 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 361) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • chemokine C10 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15807 WO0042071 Members of this family are involved in a similarly diverse range of pathologies SEQ ID NO: 49 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The WO0042071 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 438) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and Inflammatory disorders Human chemokine CCR-2 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • C-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G- type 2 protein coupled receptors. Over 40 human chemokines have been described, which (isoform A) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined (SEQ ID NO: 362) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C.
  • Human chemokine ENA- Chemokines are a family of related small, secreted proteins involved in biological 78 GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15809 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P42830/CXCL5_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-X-C motif chemokine 5 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 352) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and Inflammatory disorders Human chemokine Chemokines are a family of related small, secreted proteins involved in biological GRObeta GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Human chemokine IP-10 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15811 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P02778/CXL10_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the C-X-C motif chemokine 10 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 242) protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and Inflammatory disorders Human chemokine Chemokines are a family of related small, secreted proteins involved in biological SDF1beta GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Human chemokine GRO Chemokines are a family of related small, secreted proteins involved in biological alpha GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B15813 WO0042071 Members of this family are involved in a similarly diverse range of pathologies P09341/GROA_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The Growth-regulated alpha chemokines exert their effects by acting on a family of seven transmembrane G- protein protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological MIP1beta GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • a human C-C chemokine Chemokines are a family of related small, secreted proteins involved in biological designated exodus processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies B07939 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the 6,096,300 chemokines exert their effects by acting on a family of seven transmembrane G- P78556/CCL20_HUMAN protein coupled receptors.
  • Chemokine activities can be determined (isoform 1) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: (SEQ ID NO: 248) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer Human chemokine Chemokines are a family of related small, secreted proteins involved in biological L105_7 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • SEQ ID NO: 4 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9856818 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 437) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A.
  • Chemokines are a family of related small, secreted proteins involved in biological L105_3 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession Y96923 U.S. Members of this family are involved in a similarly diverse range of pathologies Pat. No. 6,084,071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G- WO9856818 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Chemotaxis, Gene Therapy, Wound healing Human secondary Chemokines are a family of related small, secreted proteins involved in biological lymphoid chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • Human non- ELR CXC Chemokines are a family of related small, secreted proteins involved in biological chemokine H174 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y96310 WO0029439 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the O14625/CXL11_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-X-C motif chemokine 11 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Human non-ELR CXC Chemokines are a family of related small, secreted proteins involved in biological chemokine IP10 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession Y96311 Members of this family are involved in a similarly diverse range of pathologies WO0029439 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P02778/CXL10_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-X-C motif chemokine 10 protein coupled receptors.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Human non-ELR CXC Chemokines are a family of related small, secreted proteins involved in biological chemokine Mig GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession Y96313 Members of this family are involved in a similarly diverse range of pathologies WO0029439 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the Q07325/CXCL9_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-X-C motif chemokine 9 protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological Ckbeta-7 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • FIG. 1 of WO0028035 chemokines exert their effects by acting on a family of seven transmembrane G- (SEQ ID NO: 439) protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • Human chemokine MIP- Chemokines are a family of related small, secreted proteins involved in biological 1alpha GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. Accession Y96281 Members of this family are involved in a similarly diverse range of pathologies WO0028035 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the P10147/CCL3_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 3 protein coupled receptors.
  • Chemokines are a family of related small, secreted proteins involved in biological Ckbeta-7 (optionally processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
  • GenSeq Members of this family are involved in a similarly diverse range of pathologies Accession Y96282 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the WO0028035 chemokines exert their effects by acting on a family of seven transmembrane G- FIG. 1 of WO0028035 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 440) bind to ⁇ 17 receptors thus far identified.
  • Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A.
  • Chemokines are a family of related small, secreted proteins involved in biological receptor CXCR3 GeneSeq processes ranging from Chemokine activities can be determined using assays known Accession Y79372 in the art: Methods in Molecular Biology, 2000, vol. 138: Soluble CXCR3 polypeptides WO0018431 may be useful for inhibiting P49682
  • isoform 1 SEQ ID NO: 240
  • chemokine like domain involved in a similarly diverse range of pathologies including inflammation, allergy, GeneSeq Accession tissue rejection, viral infection, and tumor biology.
  • the chemokines exert their effects Y53259 U.S. Pat. No. by acting on a family of seven transmembrane G-protein coupled receptors. Over 40 6,043,086 human chemokines have been described, which bind to ⁇ 17 receptors thus far P78423/X3CL1_HUMAN identified.
  • Chemokine Protocols Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Fractalkine Humana Press Inc., Totowa, NJ. chemokine activities and viral infection.
  • Chemokines are a family of related small, secreted proteins involved in biological chemokine interleukin C processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies Y57771 JP11302298 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The chemokines exert their effects by acting on a family of seven transmembrane G- protein coupled receptors. Over 40 human chemokines have been described, which bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol.
  • Chemokine Protocols Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ. Cancer and infectious diseases Human CKbeta-9 Chemokines are a family of related small, secreted proteins involved in biological GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking. B50860 U.S. Pat. No. Members of this family are involved in a similarly diverse range of pathologies 6,153,441 including inflammation, allergy, tissue rejection, viral infection, and tumor biology.
  • the O00585/CCL21_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G- C-C motif chemokine 21 protein coupled receptors. Over 40 human chemokines have been described, which (SEQ ID NO: 346) bind to ⁇ 17 receptors thus far identified. Chemokine activities can be determined using assays known in the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
  • Preproapolipo- protein Apoa-1 participates in the reverse transport of cholesterol from tissues to the liver for “paris” variant GeneSeq excretion by promoting cholesterol efflux from tissues and by acting as a cofactor for Accession W08602 the lecithin cholesterol acyltransferase (Icat).
  • Lipid binding activity can be determined WO9637608 using assays known in the art, such as, for example, the Cholesterol Efflux Assays of (SEQ ID NO: 466) Takahaski et al., P.N.A.S., Vol. 96, Issue 20, 11358-11363, Sep. 28, 1999.
  • Preproapolipo- protein Apoa-1 participates in the reverse transport of cholesterol from tissues to the liver for “milano” variant 5,721,114 excretion by promoting cholesterol efflux from tissues and by acting as a cofactor for SEQ ID NO: 6 of U.S. the lecithin cholesterol acyltransferase (Icat).
  • Lipid binding activity can be determined Pat. No. 5,721,114 using assays known in the art, such as, for example, the Cholesterol Efflux Assays of (SEQ ID NO: 441) Takahaski et al., P.N.A.S., Vol. 96, Issue 20, 11358-11363, Sep. 28, 1999.
  • Useful for cardio- vascular disorders, cholesterol disorders, and Hyperlipidaemia Glycodelin-A are useful for cardio- vascular disorders, cholesterol disorders, and Hyperlipidaemia Glycodelin-A; Naturally produced female contraceptive that is removed rapidly from the body Progesterone- associated following 2-3 days production. Uses include contraception Glycodelin-A activity can endometrial protein be determined using the hemizona assay as described in Oehninger, S., Coddington, C. C., GeneSeq Accession Hodgen, G. D., and Seppala, M (1995) Fertil. Steril. 63, 377-383. Naturally W00289 WO9628169 derived contraceptive useful for the prevention of pregnancy.
  • NOGO-A Genbank NOGO polypeptides are potent inhibitors of neurite growth. Inhibition of Neurite Accession CAB99248 outgrowth. Antagonists to NOGO polypeptides may promote the outgrowth of (SEQ ID NO: 366) neurites, thus inducing regeneration of neurons.
  • NOGO-A polypep- tide antagonists are useful for the pro- motion of neural growth, which could be useful in the treatment of neural disorders and dys- function due to de- generative diseases or trauma; useful in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons or to promote the structural plasticity of the CNS.
  • NOGO-B Genbank NOGO polypeptides are potent inhibitors of neurite growth. Inhibition of Neurite Accession CAB99249 outgrowth. Antagonists to NOGO polypeptides may promote the outgrowth of (SEQ ID NO: 367) neurites, thus inducing regeneration of neurons.
  • NOGO-B polypep- tide antagonists are useful for the pro- motion of neural growth, which could be useful in the treatment of neural disorders and dys- function due to de- generative diseases or trauma; useful in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons or to promote the structural plasticity of the CNS.
  • NOGO-C Genbank NOGO polypeptides are potent inhibitors of neurite growth. Inhibition of Neurite Accession CAB99250 outgrowth.
  • Antagonists to NOGO polypeptides may promote the outgrowth of (SEQ ID NO: 368) neurites, thus inducing regeneration of neurons.
  • NOGO-C polypep- tide antagonists are useful for the pro- motion of neural growth, which could be useful in the treatment of neural disorders and dys- function due to de- generative diseases or trauma; useful in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons or to promote the structural plasticity of the CNS.
  • NOGO-66 Receptor NOGO polypeptides are potent inhibitors of neurite growth, and are thought to Genbank Accession mediate their effects through the NOGO-66 Receptor. Inhibition of Neurite outgrowth AAG53612 by mediating the biological effects of NOGO polypeptides.
  • Soluble NOGO- 66 (SEQ ID NO: 369) receptor polypeptides may promote the outgrowth of neurites, thus inducing regeneration of neurons.
  • NOGO-66 receptor polypeptides are useful for the promotion of neural growth, which could be useful in the treatment of neural disorders and dys- function due to de- generative diseases or trauma; useful in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons or to promote the structural plasticity of the CNS.
  • Antibodies specific for These antibodies are useful for the promotion of neurite outgrowth Collapsin activity, collapsin U.S. Pat. No.
  • VEGF activity can Antibodies, and fragments be determined using assays known in the art, such as those disclosed in International thereof WO9845331 Publication No. WO0045835, for example. Promotion of growth and proliferation of cells, such as vascular endothelial cells. Antagonists may be useful as anti- angiogenic agents, and may be applicable for cancer Humanized Anti-VEGF These agents have anti-inflammatory and anti-cancer applications VEGF activity can Antibodies, and fragments be determined using assays known in the art, such as those disclosed in International thereof WO0029584 Publication No. WO0045835, for example.
  • Antagonists may be useful as anti-angiogenic agents, and may be applicable for cancer Membrane bound proteins Cancer, Immune Disorders
  • Cancer, Immune Disorders These proteins can be used for linking bioactive GeneSeq. Accession molecules to cells and for modulating biological activities of cells, using the Y66631-Y66765 polypeptides for specific targeting.
  • the polypeptide targeting can be used to kill the WO9963088 target cells, e.g. for the treatment of cancers.
  • Activities can be determined using assay known in the art, such as, for example, the assays disclosed in International Publication No. WO0121658.
  • proteins can be used for linking bioactive Transmembrane molecules to cells and for modulating biological activities of cells, using the polypeptides GeneSeq polypeptides for specific targeting.
  • the polypeptide targeting can be used to kill the Accession B44241-B44334 target cells, e.g. for the treatment of cancers.
  • These proteins are useful for the WO0053756 treatment of immune system disorders. Activities can be determined using assay known in the art, such as, for example, the assays disclosed in International Publication No. WO0121658.
  • proteins can be used for linking bioactive Transmembrane molecules to cells and for modulating biological activities of cells, using the polypeptides GeneSeq polypeptides for specific targeting.
  • the polypeptide targeting can be used to kill the Accession Y41685-Y41774 target cells, e.g. for the treatment of cancers.
  • These proteins are useful for the WO9946281 treatment of immune system disorders. Activities can be determined using assay known in the art, such as, for example, the assays disclosed in International Publication No. WO0121658.
  • Interleukin 2 (IL-2) Metabolic Disease Type 1 diabetes, Graft-versus-host disease.
  • IL15_vE SEQ ID NO: 553 Interleukin 15_vF Obesity, Metabolic Disease, Diabetes.
  • IL15_vF SEQ ID NO: 565 Interleukin 22 Metabolic Disease, Diabetic Ulcers, Inflamatory Bowel Diseases.
  • IL22 SEQ ID NO: 554 Fibroblast Growth Factor 1 Diabetes, Metabolic Disease, Obesity.
  • FGF1 SEQ ID NO: 555 Fibroblast Growth Factor Diabetes, Metabolic Disease, Obesity. 1_vA See Nature 513, 436-439 (18 Sep. 2014) doi: 10.1038/nature13540.
  • FGF1_vA SEQ ID NO: 556 Fibroblast Growth Factor Diabetes, Metabolic Disease, Obesity.
  • FGF1_vB See Proc Natl Acad Sci USA. 1991 Apr. 1; 88(7): 2893-2897.
  • FGF1_vB SEQ ID NO: 557 Fibroblast Growth Factor Diabetes, Metabolic Disease, Obesity.
  • FGF1_vC SEQ ID NO: 566 Fibroblast Growth Factor Chronic liver disease, primary biliary cirrhosis, bile acid-induced liver damage.
  • 19_vA See Cancer Res. 2014 Jun. 15; 74(12): 3306-16. doi: 10.1158/0008-5472.CAN-14- (FGF19_vA) 0208. Epub 2014 Apr. 11. Regulates bile acid metabolism without tumorigenicity.
  • Fibroblast Growth Factor Metabolic Disease Fibrotic Diseases. 21 (FGF21) SEQ ID NO: 559 Fibroblast Growth Factor Hyperphosphatemic familial tumoral calcinosis. 23 (FGF23) SEQ ID NO: 560 Brain-Derived Neurological diseases (including Alzheimer's Disease, Autism, Huntington's Disease, Neurotrophic Factor Parkinson's Disease, and Depression), obesity, metabolic disease.
  • BDNF Brain-Derived Neurological diseases (including Alzheimer's Disease, Autism, Huntington's Disease, Neurotrophic Factor Parkinson's Disease, and Depression), obesity, metabolic disease.
  • BDNF SEQ ID NO: 561 Serpin Family A Member 1 alpha-1-antitrypsin deficiency.
  • SERPINA1 SEQ ID NO: 584 SEQ ID NO: 585 Serpin Peptidase Inhibitor, Diabetes.
  • Clade B Optalbumin, Member 1 (SERPINB1) SEQ ID NO: 562 CASPASE1 Diabetes.
  • the nucleic acid drug including synthetic RNA
  • is administered is a manner that it effects one or more of keratinocytes and fibroblasts (e.g. causes these cells to express one or more therapeutic proteins).
  • present methods allow for methods in which a patient's cells are used to generate a therapeutic protein and the levels of such protein are tailored by synthetic RNA dosing.
  • the synthetic RNA targets a soluble protein.
  • the synthetic RNA targets a protein of one or more of the following families of proteins: transforming growth factor (TGF) beta, bone morphogenetic proteins (BMPs), Fibroblast growth factors (FGFs), vascular endothelial growth factors (VEGFs), and interleukins.
  • TGF transforming growth factor
  • BMPs bone morphogenetic proteins
  • FGFs Fibroblast growth factors
  • VEGFs vascular endothelial growth factors
  • interleukins interleukins.
  • the synthetic RNA targets a member of the TGF beta family.
  • TGF- ⁇ superfamily proteins comprise cytokines characterized by six-conserved cysteine residues (Lander et al., (2001) Nature, 409:860-921). The human genome contains at least about 42 open reading frames encoding TGF- ⁇ superfamily proteins.
  • TGF- ⁇ superfamily proteins can at least be divided into the BMP subfamily and the TGF- ⁇ subfamily based on sequence similarity and the specific signaling pathways that they activate.
  • the synthetic RNA targets one or more of TGFs (e.g., TGF- ⁇ 1, TGF- ⁇ 2, and TGF- ⁇ 3), activins (e.g., activin A) and inhibins, macrophage inhibitory cytokine-1 (MIC-1), Mullerian inhibiting substance, anti-Mullerian hormone, and glial cell line derived neurotrophic factor (GDNF).
  • TGFs e.g., TGF- ⁇ 1, TGF- ⁇ 2, and TGF- ⁇ 3
  • activins e.g., activin A
  • inhibins e.g., macrophage inhibitory cytokine-1 (MIC-1), Mullerian inhibiting substance, anti-Mullerian hormone, and glial cell line derived neurotrophic factor (GDNF).
  • TGFs e.g., TGF- ⁇ 1, TGF- ⁇ 2, and TGF- ⁇ 3
  • activins e.g., activin A
  • MIC-1 macrophage inhibitory cytokine-1
  • the TGF- ⁇ superfamily comprises a subset of the cysteine knot cytokine superfamily. Additional members of the cysteine knot cytokine superfamily include, but are not limited to, platelet derived growth factor (PDGF), vascular endothelial growth factor (VEGF), placenta growth factor (P1GF), Noggin, neurotrophins (BDNF, NT3, NT4, and PNGF), gonadotropin, follitropin, lutropin, interleukin-17, and coagulogen. This family of proteins is also among the targets encompassed by the present invention.
  • PDGF platelet derived growth factor
  • VEGF vascular endothelial growth factor
  • P1GF placenta growth factor
  • Noggin neurotrophins
  • BDNF, NT3, NT4, and PNGF neurotrophins
  • gonadotropin gonadotropin
  • follitropin lutropin
  • interleukin-17 interleukin-17
  • coagulogen
  • the present invention relates to targeting a TGF beta family member for treatment or prevention of various immunological disorders, cancer, bronchial asthma, lung fibrosis, heart disease, diabetes, hereditary hemorrhagic telangiectasia, Marfan syndrome, Vascular Ehlers-Danlos syndrome, Loeys-Dietz syndrome, Parkinson's disease, chronic kidney disease, multiple sclerosis and AIDS.
  • the synthetic RNA targets a member of the BMP family.
  • the BMP subfamily includes, but is not limited to, BMP-2, BMP-3 (osteogenin), BMP-3b (GDF-10), BMP-4 (BMP-2b), BMP-5, BMP-6, BMP-7 (osteogenic protein-1 or OP-1), BMP-8 (OP-2), BMP-8B (OP-3), BMP-9 (GDF-2), BMP-10, BMP-11 (GDF-11), BMP-12 (GDF-7), BMP-13 (GDF-6, CDMP-2), BMP-15 (GDF-9), BMP-16, GDF-1, GDF-3, GDF-5 (CDMP-1), and GDF-8 (myostatin).
  • the synthetic RNA targets one or more of BMP-2, BMP-3 (osteogenin), BMP-3b (GDF-10), BMP-4 (BMP-2b), BMP-5, BMP-6, BMP-7 (osteogenic protein-1 or OP-1), BMP-8 (OP-2), BMP-8B (OP-3), BMP-9 (GDF-2), BMP-10, BMP-11 (GDF-11), BMP-12 (GDF-7), BMP-13 (GDF-6, CDMP-2), BMP-15 (GDF-9), BMP-16, GDF-1, GDF-3, GDF-5 (CDMP-1), and GDF-8 (myostatin).
  • BMP-2 BMP-3 (osteogenin), BMP-3b (GDF-10), BMP-4 (BMP-2b), BMP-5, BMP-6, BMP-7 (osteogenic protein-1 or OP-1), BMP-8 (OP-2), BMP-8B (OP-3), BMP-9 (GDF-2), BMP-10, BMP-11 (GDF-11), BMP-12 (GDF-7), BMP
  • BMPs are sometimes referred to as Osteogenic Protein (OPs), Growth Differentiation Factors (GDFs), or Cartilage-Derived Morphogenetic Proteins (CDMPs).
  • the synthetic RNA targets one or more BMP fusions (e.g. as described in US Patent Publication No. 2009/0202638, the entire contents of which are hereby incorporated by reference) and/or one or more BMP mutants (e.g. as described in US Patent Publication No. 2011/0039773, the entire contents of which are hereby incorporated by reference).
  • the present invention relates to targeting a BMP family member for regenerative medicine or metabolic applications, including without limitation orthopedic applications such as spinal fusions, nonunions and oral surgerie, metabolic disease, prediabetes, diabetes, thermogenesis, insulin sensitivity, insulin resistance, and adipogenesis, including brown-fat adipogenesis.
  • orthopedic applications such as spinal fusions, nonunions and oral surgerie
  • metabolic disease prediabetes
  • diabetes thermogenesis
  • insulin sensitivity insulin resistance
  • adipogenesis including brown-fat adipogenesis.
  • adipogenesis including brown-fat adipogenesis.
  • the present invention relates to targeting a BMP family member, including BMP-7, for treatment of chronic kidney disease (CKD) and/or to reverse the loss of glomeruli due to sclerosis.
  • CKD chronic kidney disease
  • the present invention relates to targeting a BMP family member to induce proliferation of bone and cartilage in a variety of locations in the body.
  • repair of joints such as knee, elbow, ankle, and finger joints are contemplated by the invention.
  • targeting a BMP family member may result in regenerating cartilage in patients suffering from arthritis or other cartilage degenerating diseases.
  • the invention pertains to treating tears in cartilage due to injury.
  • the invention is useful for inducing bone growth in patients, for instance, by way of non-limitation, for use in treating patients suffering from bone fractures or breaks, osteoporosis, or patients in need of spinal fusion or for repair of the spine, vertebrae or the like.
  • the present invention relates to targeting a BMP family member to induce a developmental cascade of bone morphogenesis and tissue morphogenesis for a variety of tissues in mammals different from bone or bone cartilage.
  • This morphogenic activity includes the ability to induce proliferation and differentiation of progenitor cells, and the ability to support and maintain the differentiated phenotype through the progression of events that results in the formation of bone, cartilage, non-mineralized skeletal or connective tissues, and other adult tissues.
  • the present invention may be used for treatment to prevent loss of and/or increase bone mass in metabolic bone diseases.
  • General methods for treatment to prevent loss of and/or increase bone mass in metabolic bone diseases using osteogenic proteins are disclosed in U.S. Pat. No. 5,674,844, the entire contents of which are hereby incorporated by reference.
  • the present compositions and methods find use in replacing or repairing bone or cartilage at injury sites such as bone breaks, bone fractures, and cartilage tears, periodontal tissue regeneration (e.g. general methods for periodontal tissue regeneration using osteogenic proteins are disclosed in U.S. Pat. No. 5,733,878, the entire contents of which are hereby incorporated by reference), liver regeneration, including following a partial hepatectomy (see, e.g., U.S. Pat. No.
  • compositions and methods including when targeting one or more BMPs, can be used to induce dentinogenesis.
  • the unpredictable response of dental pulp tissue to injury is a basic clinical problem in dentistry.
  • small areas e.g., 2 mm
  • dental pulps can be surgically exposed by removing the enamel and dentin immediately above the pulp (by drilling) of sample teeth, performing a partial amputation of the coronal pulp tissue, inducing hemostasis, application of the pulp treatment, and sealing and filling the cavity by standard procedures.
  • the present invention relates to targeting a BMP family member for the treatment of one or more metabolic-related disorders as described herein.
  • the synthetic RNA targets a member of the FGF family.
  • FGFs are a family of growth factors, with members involved in angiogenesis, wound healing, embryonic development and various endocrine signaling pathways.
  • any one of the following FGFs are targets of the invention: FGF1, FGF2, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, FGF10, FGF11, FGF12, FGF13, FGF14 (FGF11, FGF12, FGF13, and FGF14 being FGF homologous factors 1-4 (FHF1-FHF4)), FGF16, FGF17, FGF18, FGF19 (aka FHF15/19), FGF20, FGF21, FGF22 and FGF22.
  • the synthetic RNA targets a cognate receptor of any of the above FGFs.
  • the present invention relates to targeting a FGF family member to a treat or prevent disease or disorder associated with abnormal function and/or expression of an FGF, a metabolic disease or disorder (e.g., diabetes, obesity, dyslipidemia, hyperglycemia, hyperinsulinemia, hypertension, hepatosteaotosis such as non-alcoholic steatohepatitis (NASH) etc.), cancer, a disease or disorder associated with impaired lipid metabolism a disease or disorder associated with impaired renal function, a disease or disorder associated with impaired hepatic function, abnormal cell proliferation, a vascular disease or disorder (e.g., coronary artery disease, peripheral artery disease, atherosclerosis, abdominal aortic aneurysm, a blood clot, deep vein thrombosis, venous stasis disease, phlebitis, varicose veins etc.), angiogenesis, athe
  • the synthetic RNA targets a member of the VEGF family.
  • the target is one or more of VEGF-A (including all isoforms, e.g. VEGF121, VEGF165 and VEGF189), placenta growth factor (PGF, including all isoforms, e.g. PGF-1, PGF-2, and PGF-3), VEGF-B, VEGF-C and VEGF-D, and any variants thereof (see, e.g. U.S. Pat. No. 9,078,860, the entire contents of which are hereby incorporated by reference).
  • the synthetic RNA targets a cognate receptor of any of the above VEGFs.
  • VEGF-related proteins including orf virus-encoded VEGF-like proteins referred to as VEGF-E and a series of snake venoms referred to as VEGF-F.
  • VEGFs and VEGF-related proteins are members of the Platelet Derived Growth Factor (PDGF) supergene family of cystine knot growth factors. All members of the PDGF supergene family share a high degree of structural homology with PDGF.
  • PDGF Platelet Derived Growth Factor
  • the present invention relates to targeting a VEGF family member to treat diseases and conditions associated with angiogenesis, including but not limited to, solid tumor cancers, hemangiomas, rheumatoid arthritis, osteoarthritis, septic arthritis, asthma, atherosclerosis, idiopathic pulmonary fibrosis, vascular restenosis, arteriovenous malformations, meningiomas, neovascular glaucoma, psoriasis, Kaposi's Syndrome, angiofibroma, hemophilic joints, hypertrophic scars, Osler-Weber syndrome, pyogenic granuloma, retrolental fibroplasias, scleroderma, trachoma, von Hippel-Lindau disease, vascular adhesion pathologies, synovitis, dermatitis, neurological degenerative diseases, preeclampsia, unexplained female infertility, endometriosis, unexplained male
  • the present invention relates to targeting a VEGF family member to treat angiogenesis-associated eye diseases, including without limitation any eye disease associated with abnormal intraocular neovascularization, including but not limited to retinopathy of prematurity, diabetic retinopathy, retinal vein occlusion, and age-related macular degeneration, as well diabetic macular edema and retinal vein occlusion.
  • angiogenesis-associated eye diseases including without limitation any eye disease associated with abnormal intraocular neovascularization, including but not limited to retinopathy of prematurity, diabetic retinopathy, retinal vein occlusion, and age-related macular degeneration, as well diabetic macular edema and retinal vein occlusion.
  • the present compositions and methods relate to the treatment of wet age-related macular degeneration.
  • the synthetic RNA targets a member of the interleukin family.
  • the interleukins represent a large group of cytokines with diverse functions and were first characterized by expression in leukocytes and have since been shown to be expressed in a wide variety of cells, for example macrophages, TH-1 and TH-2 cells, T-lymphocytes, monocytes and bone marrow stroma. Broadly, the function of the immune system depends in a large part on the expression and function of the interleukins.
  • the target is one or more of interleukins 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35 and 36, and, within each species of interleukin, various isotypes and/or interleukin receptors (e.g., IL-1R, IL-2R, IL-3R, IL-4R, IL-5R, IL-6R, IL-7R, IL-8R, IL-9R, IL-10R, IL-11R, IL-12R, IL-13R, IL-14R, IL-15R, IL-16R, IL-17R, IL-18R, IL-19R, IL-20R, IL-21R, IL-22R, IL-23R, IL-24R, IL-25R, IL-26, IL-27R, IL-28R, IL-29R, IL-30R, IL-31R, IL-32R, IL-
  • both IL-15 and IL-15R are targeted.
  • the present invention relates to targeting a member of the interleukin family to treat cancer, inflammatory, respiratory, autoimmune, cardiovascular, neurological, metabolic, and/or proliferative diseases, disorders, and/or conditions in a subject or organism, as described herein.
  • the present invention relates to targeting a member of the interleukin family to treat cancer.
  • the present invention relates to targeting a member of the interleukin family to treat rheumatoid arthritis.
  • the synthetic RNA targets an EPO gene or a derivative thereof (e.g. SEQ ID NO: 164, SEQ ID NO: 165, SEQ ID NO: 166, and SEQ ID NO: 167).
  • EPO gene or a derivative thereof e.g. SEQ ID NO: 164, SEQ ID NO: 165, SEQ ID NO: 166, and SEQ ID NO: 167.
  • Some embodiments are related to NOVEPOETIN protein (SEQ ID NO: 167).
  • Other embodiments are related to NOVECRIT (SEQ ID NO: 168).
  • Erythropoietin can stimulate erythropoiesis in anemic patients with chronic renal failure in whom the endogenous production of erythropoietin is impaired. Without being bound by theory, because of the length of time often required for erythropoiesis (several days for erythroid progenitors to mature and be released into the circulation), a clinically significant increase in hemoglobin is usually not observed in less than two weeks and may require up to ten weeks in some patients. The present methods and compositions provide, in some embodiments, more rapid therapeutic effect.
  • the present methods and compositions provide, in some embodiments, more rapid therapeutic effect, for instance, when compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic.
  • the present methods and compositions provide, in some embodiments, a sustained therapeutic effect.
  • the present methods and compositions provide, in some embodiments, a sustained therapeutic effect, for instance, when compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic.
  • the present methods and compositions provide a clinically significant increase in hematocrit in less than about 6 weeks, or less than about 5 weeks, or less than about 4 weeks, or less than about 3 weeks, or less than about 2 weeks, or less than about 1 week.
  • the present methods and compositions provide a clinically significant increase in hematocrit in about 2 weeks, or about 10 days, or about 1 week, or about 3 days, or about 1 day.
  • the present methods and compositions accelerate the process by which erythroid progenitors mature and are released into the circulation.
  • the present EPO-related compositions find use in decreasing the dose and/or frequency of administration when compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic.
  • the present EPO-related compositions may find use in treatment regimens for the diseases disclosed herein (including, without limitation, one or more anemias) that involve administration on a monthly, or biweekly, or weekly basis. In some embodiments, therefore, the present EPO-related compositions reduce the need for daily, or, in some embodiments, weekly, administration.
  • the present EPO-related compositions require lower maintenance doses as compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic. Certain embodiments are particularly useful for treating chemotherapy-induced anemia. Other embodiments are particularly useful for treating anemia associated with inflammation, including, but not limited to, rheumatoid arthritis.
  • the present the present methods and compositions provide a fast and robust response that obviates the need for RBC transfusion.
  • the present methods and compositions allow for treatment of patients do not consent to transfusions.
  • the present methods and compositions increase the rate of increase in hematocrit. In some embodiments, the present methods and compositions maintain elevated hematocrits (e.g. of 25%, or 30%, or 35%, or 40% or more) for a sustained period (e.g. about 1 month, or about 2 months, or about 3 months, or about 4 months, or about 5 months, or about 6 months, or about 9 months).
  • elevated hematocrits e.g. of 25%, or 30%, or 35%, or 40% or more
  • a sustained period e.g. about 1 month, or about 2 months, or about 3 months, or about 4 months, or about 5 months, or about 6 months, or about 9 months.
  • the present methods and compositions stimulate red blood cell production. In some embodiments, the present methods and compositions stimulate division and differentiation of committed erythroid progenitors in the bone marrow.
  • the present invention relates to the treatment of one or more of anemia, including anemia resulting from resulting from chronic kidney disease (e.g. from dialysis) and/or chemotherapy and/or HIV treatment (e.g. Zidovudine (INN) or azidothymidine (AZT)), inflammatory bowel disease (e.g. Crohn's disease and ulcer colitis), anemia linked to inflammatory conditions (e.g. arthritis, lupus, IBD), anemia linked to diabetes, schizophrenia, cerebral malaria, as aplastic anemia, and myelodysplasia from the treatment of cancer (e.g. chemotherapy and/or radiation), and various myelodysplastic syndrome diseases (e.g. sickle cell anemia, hemoglobin SC disease, hemoglobin C disease, alpha- and beta-thalassemias, neonatal anemia after premature birth, and comparable conditions).
  • anemia including anemia resulting from resulting from chronic kidney disease (e.g. from dialysis) and/or chemotherapy and/or HIV treatment (e
  • the present invention relates to the treatment of, or a patient having one or more of cancer, heart failure, autoimmune disease, sickle cell disease, thalassemia, blood loss, transfusion reaction, diabetes, vitamin B12 deficiency, collagen vascular disease, Shwachman syndrome, thrombocytopenic purpura, Celiac disease, endocrine deficiency state such as hypothyroidism or Addison's disease, autoimmune disease such as Crohn's Disease, systemic lupus erythematosis, rheumatoid arthritis or juvenile rheumatoid arthritis, ulcerative colitis immune disorders such as eosinophilic fasciitis, hypoimmunoglobulinemia, or thymoma/thymic carcinoma, graft vs.
  • cancer heart failure
  • autoimmune disease sickle cell disease
  • thalassemia blood loss
  • transfusion reaction diabetes
  • vitamin B12 deficiency collagen vascular disease
  • Shwachman syndrome thrombocytopen
  • hematologic syndrome e.g. Down's, Dubowwitz, Seckel
  • Felty syndrome hemolytic uremic syndrome
  • myelodysplasic syndrome nocturnal paroxysmal hemoglobinuria
  • osteomyelofibrosis pancytopenia, pure red-cell aplasia
  • Schoenlein-Henoch purpura malaria
  • protein starvation menorrhagia
  • systemic sclerosis liver cirrhosis
  • hypometabolic states congestive heart failure
  • chronic infections such as HIV/AIDS, tuberculosis, oseomyelitis, hepatitis B, hepatitis C, Epstein-barr virus or parvovirus
  • T cell leukemia virus bacterial overgrowth syndrome
  • fungal or parasitic infections and/or red cell membrane disorders
  • red cell membrane disorders such as hereditary spherocytosis, hereditary elliptocytosis, heriditray pyrpoikilocytosis
  • the present invention relates to the treatment of, or a patient having anemia, i.e. a condition in which the number of red blood cells and/or the amount of hemoglobin found in the red blood cells is below normal.
  • anemia i.e. a condition in which the number of red blood cells and/or the amount of hemoglobin found in the red blood cells is below normal.
  • the anemia may be acute or chronic.
  • the present anemias include but are not limited to iron deficiency anemia, renal anemia, anemia of chronic diseases/inflammation, pernicious anemia such as macrocytic achylic anemia, juvenile pernicious anemia and congenital pernicious anemia, cancer-related anemia, chemotherapy-related anemia, radiotherapy-related anemia, pure red cell aplasia, refractory anemia with excess of blasts, aplastic anemia, X-lined siderobalstic anemia, hemolytic anemia, sickle cell anemia, anemia caused by impaired production of ESA, myelodysplasia syndromes, hypochromic anemia, microcytic anemia, sideroblastic anemia, autoimmune hemolytic anemia, Cooley's anemia, Mediterranean anemia, Diamond Blackfan anemia, Fanconi's anemia and drug-induced immune hemolytic anemia.
  • Anemia may cause serious symptoms, including hypoxia, chronic fatigue, lack of concentration, pale skin, low blood pressure, dizziness and heart failure.
  • the anemia is induced by chemotherapy is one cause of anemia.
  • the chemotherapy may be any myelosuppressive chemotherapy.
  • the chemotherapy is one or more platinum-based drugs including cisplatin (e.g. PLATINOL) and carboplatin (e.g. PARAPLATIN).
  • the chemotherapy is any one of the agents described herein.
  • the chemotherapy is any agent described in Groopman et al. J Natl Cancer Inst (1999) 91 (19): 1616-1634, the contents of which are hereby incorporated by reference in their entireties.
  • the present compositions and methods are used in the treatment of chemotherapy-related anemia in later stage cancer patients (e.g. a stage IV, or stage III, or stage II cancer).
  • the present compositions and methods are used in the treatment of chemotherapy-related anemia in cancer patients receiving dose-dense chemotherapy or other aggressive chemotherapy regimens.
  • the present invention relates to the treatment of anemia in a patient having one or more blood-based cancers, such as leukemia, lymphoma, and multiple myeloma. Such cancers may affect the bone marrow directly. Further, the present invention relates to metastatic cancer that has spread to the bone or bone marrow. In some embodiments, the present invention relates to the treatment of anemia in a patient undergoing radiation therapy. Such radiation therapy may damage the bone marrow, lowering its ability to make red blood cells. In further embodiments, the present invention relates to the treatment of anemia in a patient having a reduction or deficiency of one or more of iron, vitamin B12, and folic acid.
  • blood-based cancers such as leukemia, lymphoma, and multiple myeloma.
  • the present invention relates to metastatic cancer that has spread to the bone or bone marrow.
  • the present invention relates to the treatment of anemia in a patient undergoing radiation therapy. Such radiation therapy may damage the bone marrow
  • the present invention relates to the treatment of anemia in a patient having excessive bleeding including without limitation, after surgery or from a tumor that is causing internal bleeding. In further embodiments, the present invention relates to the treatment of anemia in a patient having anemia of chronic disease.
  • the present invention relates to the treatment of anemia resulting from chronic renal failure. In some embodiments, the present invention relates to the treatment of anemia resulting from the use of one or more renal replacement therapies, inclusive of dialysis, hemodialysis, peritoneal dialysis, hemofiltration, hemodiafiltration, and renal transplantation.
  • the present invention relates to the treatment of anemia in patients with chronic kidney disease who are not on dialysis.
  • the present invention relates to patients in stage 1 CKD, or stage 2 CKD, or stage 3 CKD, or stage 4 CKD, or stage 5 CKD.
  • the present patient is stage 4 CKD or stage 5 CKD.
  • the present patient has undergone a kidney transplant.
  • the present invention relates to the treatment of anemia is a patient having an acute kidney injury (AKI).
  • AKI acute kidney injury
  • compositions and methods are used to reduce or eliminate fatigue, dizziness, and shortness of breath in a patient.
  • hyporesponsiveness to erythropoietin or ESA-resistant anemia refers to the presence of at least one of the following conditions: i) a significant decrease in hemoglobin levels at a constant dose of ESA treatment, ii) a significant increase in the ESA dose requirement to achieve or maintain a certain hemoglobin level, iii) a failure to raise the hemoglobin level to the target range despite the ESA dose equivalent to erythropoietin greater than 150 IU/kg/week or 0.75 mg/kg/week of darbepoeitn-alpha or continued need for such high dose of ESA to maintain the target hemoglobin level.
  • the present compositions and methods mitigate the need for dose-escalation of the erythropoiesis stimulating agent therapy and therefore, optionally, avoid side effects (e.g. flu-like symptoms such as joint pains, weakness, dizziness and tiredness, skin irritation, increased risk of adverse cardiovascular complications).
  • side effects e.g. flu-like symptoms such as joint pains, weakness, dizziness and tiredness, skin irritation, increased risk of adverse cardiovascular complications.
  • the present compositions and methods are used to maintain a hemoglobin level of about 12.5 to 13 g/dL. In various embodiments, the present compositions and methods are used in patients having hemoglobin levels of below about 12 g/dL, or about 11 g/dL, or about 10 g/dL, or about 9 g/dL, or about 8 g/dL, or about 7 g/dL, or about 6 g/dL, or about 5 g/dL. In various embodiments, the present compositions and methods are used in patients having iron blood test scores that indicate blood pathology, e.g. a ferritin score of below about 200 ng/L and/or a transferrin saturation score below about 30%.
  • iron blood test scores that indicate blood pathology
  • the present compositions and methods are used to increase or maintain hemoglobin levels at a target level ranging from 9 to 10 g/dL, at a target level ranging from 9 g/dL to 11 g/dL, at a target level ranging from 9 g/dL to 12 g/dL, at a target level ranging from 9 g/dL to 14 g/dL, at a target level ranging from 10 g/dL to 14 g/dL, or at a target level ranging from 12 g/dL to 14 g/dL.
  • the present compositions and methods are used to bring a patient's hemoglobin levels to normal.
  • normal hemoglobin ranges for humans are about 14-18 g/dl for men and 12-16 for women g/dl with the average hemoglobin value for men at about 16 g/dL and for women at about 14 g/dL.
  • the present invention relates to the treatment of anemia of one or more of the following toxicity grading criteria (e.g. NCI Common Toxicity Criteria): grade 1 (mild), 10.0 g hemoglobin/dL to within normal limits; grade 2 (moderate), 8.0-10.0 g of hemoglobin/dL; grade 3 (serious or severe), 6.5-7.9 g of hemoglobin/dL; and grade 4 (life threatening), less than 6.5 g of hemoglobin/dL.
  • the present invention brings an increase in toxicity grading criteria by about 1 point, or about 2 points, or about 3 points, or about 4 points.
  • the present invention results in a patient having a level of 0 or 1.
  • the present compositions and methods improve anemia as assessed by one or more scales described in Groopman et al. J Natl Cancer Inst (1999) 91 (19): 1616-1634, the entire contents of which are hereby incorporated by reference in their entireties.
  • the present invention when targeting EPO, relates to combination therapy with one or more EPOs.
  • the present compositions may provide a sustained effect that can supplement a fast action of another EPO.
  • the present compositions are used as an adjuvant to other EPOs.
  • the present compositions are used as a maintenance therapy to other EPOs
  • EPOs include the following: epoetin alfa, including without limitation, DARBEPOETIN (ARANESP), EPOCEPT (LUPIN PHARMA), NANOKINE (NANOGEN PHARMACEUTICAL), EPOFIT (INTAS PHARMA), EPOGEN (AMGEN), EPOGIN, EPREX, (JANSSEN-CILAG), BINOCRIT (SANDOZ), PROCRIT; epoetin beta, including without limitation, NEORECORMON (HOFFMANN-LA ROCHE), RECORMON, Methoxy polyethylene glycol-epoetin beta (MIRCERA, ROCHE); epoetin delta, including without limitation, DYNEPO (erythropoiesis stimulating protein, SHIRE PLC); epoetin omega, including without limitation, EPOMAX; epoetin zeta, including without limitation, SILAPO (ST
  • the present invention when targeting EPO, relates to combination therapy with a blood transfusion.
  • the present compositions may supplement a blood transfusion.
  • the present invention when targeting EPO, relates to combination therapy with iron supplements.
  • the present invention also relates to the following protein targets, e.g. in the treatment of disease: growth hormone (GH) e.g. human and bovine growth hormone, growth hormone-releasing hormones; interferon including ⁇ -, ⁇ -, or ⁇ -interferons, etc., interleukin-I; interleukin-II; erythropoietin including ⁇ - and ⁇ -erythropoietin (EPO), granulocyte colony stimulating factor (GCSF), granulocyte macrophage colony stimulating factor (GM-CSF), anti-agiogenic proteins (e.g., angiostatin, endostatin) PACAP polypeptide (pituitary adenylate cyclase activating polypeptide), vasoactive intestinal peptide (VIP), thyrotrophin releasing hormone (TRH), corticotropin releasing hormone (CRH), vasopressin, arginine vasopressin (AVP), angioten
  • GH
  • the present methods allow for effective additional therapeutic agent (e.g. those described herein) activity and/or targeting to a cell and/or tissue of interest.
  • the present synthetic RNA can lead to increased expression of one or more targeting molecules that direct an additional therapeutic to the location of therapy.
  • the additional therapeutic agent may have a binding partner that the synthetic RNA encodes.
  • the synthetic RNA may induce the expression of an antigen that directs the therapeutic activity of an antibody that may be used in combination (e.g.
  • the synthetic RNA can be injected directly into one or more of the tumors described herein and home the therapeutic antibody to the tumor.
  • the present methods allow for effective additional therapeutic agent generation, especially when the additional therapeutic agent is a prodrug, for example, to produce an active form of the drug.
  • the synthetic RNA can be injected directly into one or more of the tumors described herein and home the prodrug to the tumor.
  • the synthetic RNA may encode an enzyme that catalyzes the localized conversion of a non-toxic, systemically delivered agent into a potent chemotherapeutic agent.
  • Enzyme Prodrug Drug aldehyde oxidases 5-Ethynyl-2(1H)-pyrimidinone 5-Ethynyluracil aldehyde oxidases IPdR IUdR aldehyde oxidases 5-FP 5-FU amino acid oxidases d-alanine Hydrogen peroxide amino acid oxidases SeCys conjugates Selenols and hydrogen peroxide cytochrome P450 reductase Menadione Semiquinone radical cytochrome P450 reductase Mitomycin C Quinone methide intermediate cytochrome P450 reductase Tirapazamine Nitroxide radical cytochrome P450 reductase EO9 Unidentified semiquinone radical DT-diaphorase Streptonigrin Unidentified DT-diaphorase Mitomycin C Quinone methide intermediate DT-diaphorase CB 1954 5-(Aziridin-1-yl)-4-hydroxyl
  • the synthetic RNA may encode an enzyme that catalyzes the conversion of 5-FU and/or Doxorubicin from various prodrugs, as illustrated by the examples below:
  • the nucleic acid drugs at the doses and regimens described herein may be used in combination with one or more additional agents (aka adjuvant therapy or combination agent).
  • the nucleic acid drugs at the doses and regimens described herein may be used in a human patient undergoing treatment with one or more additional agents.
  • the nucleic acid drug is used as an adjuvant or neoadjuvant to any of the additional agents described herein.
  • the invention pertains to co-administration and/or co-formulation. Any of the compositions described herein may be co-formulated and/or co-administered.
  • any nucleic acid drug described herein acts synergistically when co-administered with another agent and may be administered at doses that are lower than the doses commonly employed when such agents are used as monotherapy.
  • any nucleic acid drug described herein may include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the composition such that covalent attachment does not prevent the activity of the composition.
  • derivatives include composition that have been modified by, inter alia, glycosylation, lipidation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications can be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of turicamycin, etc. Additionally, the derivative can contain one or more non-classical amino acids. In various embodiments, one or more additional agents (aka adjuvant therapy or combination agent) may be conjugated to any nucleic acid drug described herein.
  • a cell with a steroid can suppress the innate immune response to foreign nucleic acids, and can increase the efficiency of nucleic acid delivery and translation. Certain embodiments are therefore directed to contacting a cell with a steroid. Other embodiments are directed to administering a steroid to a patient.
  • Illustrative steroids include corticosteroid steriods.
  • the steroid is one or more of cortisone, hydrocortisone, prednisone, prednisolone, dexamethasone, triamcinolone, and betamethasone.
  • the steroid is hydrocortisone.
  • the steroid is dexamethasone.
  • inventions are directed to administering to a patient a member of the group: an antibiotic, an antimycotic, and an RNAse inhibitor.
  • Botulinum toxin type A has been approved by the US Food and Drug Administration (FDA) for the treatment of essential blepharospasm, strabismus and hemifacial spasm in patients over the age of twelve, cervical dystonia, glabellar line (facial) wrinkles and for treating hyperhydrosis and botulinum toxin type B has been approved for the treatment of cervical dystonia.
  • FDA US Food and Drug Administration
  • the present compositions may be combined with these toxins in the treatment of these diseases and related diseases.
  • Some embodiments are directed to a nucleic acid drug targeting a neurotoxin.
  • the neurotoxin is a botulinum toxin or a biologically active fragment, variant, analogue or family-member thereof.
  • any one of the aforementioned toxins may be used in combination with the present compositions for various cosmetic procedures, including, without limitation, facial wrinkles, hyperkinetic skin lines, glabellar lines, crow's feet, marionette lines, skin disorders, nasolabial folds, blepharospasm, strabismus, hemifacial spasms and sweating disorders.
  • the present compositions may be used to in these cosmetic procedures as a monotherapy.
  • Certain embodiments are directed to a combination therapy comprising one or more of the therapeutic or cosmetic compositions of the present invention and one or more adjuvant therapies or cosmetic treatments.
  • one or more of the therapeutic or cosmetic compositions of the present invention are administered to a subject which is undergoing treatment with one or more adjuvant therapies or cosmetic treatments.
  • Example adjuvant therapies and cosmetic treatments are set forth in Table 3 and Table 5 of U.S. Provisional Application No. 61/721,302, the contents of which are hereby incorporated by reference, and are given by way of example, and not by way of limitation.
  • the additional agent is a cytotoxic agent, comprising, in illustrative embodiments, a toxin, a chemotherapeutic agent, a radioisotope, and an agent that causes apoptosis or cell death.
  • Illustrative cytotoxic agents include, but are not limited to, methotrexate, aminopterin, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine; alkylating agents such as mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU), mitomycin C, lomustine (CCNU), 1-methylnitrosourea, cyclothosphamide, mechlorethamine, busulfan, dibromomannitol, streptozotocin, mitomycin C, cis-dichlorodiamine platinum (II) (DDP) cisplatin and carboplatin (paraplatin); anthracyclines include daunorubicin (formerly daunomycin), doxorubicin (adriamycin), detorubicin, carminomycin, idarubicin, epirubicin, mitoxantron
  • cytotoxic agents include paclitaxel (taxol), ricin, pseudomonas exotoxin, gemcitabine, cytochalasin B, gramicidin D, ethidium bromide, emetine, etoposide, tenoposide, colchicin, dihydroxy anthracin dione, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, procarbazine, hydroxyurea, asparaginase, corticosteroids, mytotane (O,P′-(DDD)), interferons, and mixtures of these cytotoxic agents.
  • taxol taxol
  • ricin pseudomonas exotoxin
  • gemcitabine cytochalasin B
  • gramicidin D ethidium bromide
  • emetine emetine
  • etoposide tenoposide
  • cytotoxic agents include, but are not limited to, chemotherapeutic agents such as carboplatin, cisplatin, paclitaxel, gemcitabine, calicheamicin, doxorubicin, 5-fluorouracil, mitomycin C, actinomycin D, cyclophosphamide, vincristine, bleomycin, VEGF antagonists, EGFR antagonists, platins, taxols, irinotecan, 5-fluorouracil, gemcytabine, leucovorine, steroids, cyclophosphamide, melphalan, vinca alkaloids (e.g., vinblastine, vincristine, vindesine and vinorelbine), mustines, tyrosine kinase inhibitors, radiotherapy, sex hormone antagonists, selective androgen receptor modulators, selective estrogen receptor modulators, PDGF antagonists, TNF antagonists, IL-1 antagonists, interleukins (e.g.
  • IL-12 or IL-2 IL-12R antagonists
  • Toxin conjugated monoclonal antibodies tumor antigen specific monoclonal antibodies
  • Erbitux Avastin
  • Pertuzumab anti-CD20 antibodies
  • Rituxan ocrelizumab
  • ofatumumab DXL625, HERCEPTIN, or any combination thereof.
  • Toxic enzymes from plants and bacteria such as ricin, diphtheria toxin and Pseudomonas toxin may be conjugated to the therapeutic agents (e.g. antibodies) to generate cell-type-specific-killing reagents (Youle, et al., Proc. Nat'l Acad. Sci.
  • cytotoxic agents include cytotoxic ribonucleases as described by Goldenberg in U.S. Pat. No. 6,653,104.
  • Embodiments of the invention also relate to radioimmunoconjugates where a radionuclide that emits alpha or beta particles is stably coupled to the antibody, or binding fragments thereof, with or without the use of a complex-forming agent.
  • radionuclides include beta-emitters such as Phosphorus-32, Scandium-47, Copper-67, Gallium-67, Yttrium-88, Yttrium-90, Iodine-125, Iodine-131, Samarium-153, Lutetium-177, Rhenium-186 or Rhenium-188, and alpha-emitters such as Astatine-211, Lead-212, Bismuth-212, Bismuth-213 or Actinium-225.
  • beta-emitters such as Phosphorus-32, Scandium-47, Copper-67, Gallium-67, Yttrium-88, Yttrium-90, Iodine-125, Iodine-131, Samarium-153, Lutetium-177, Rhenium-186 or Rhenium-188
  • alpha-emitters such as Astatine-211, Lead-212, Bismuth-212, Bismuth-213 or Actinium-225.
  • Illustrative detectable moieties further include, but are not limited to, horseradish peroxidase, acetylcholinesterase, alkaline phosphatase, beta-galactosidase and luciferase.
  • Further exemplary fluorescent materials include, but are not limited to, rhodamine, fluorescein, fluorescein isothiocyanate, umbelliferone, dichlorotriazinylamine, phycoerythrin and dansyl chloride.
  • Further chemiluminescent moieties include, but are not limited to, luminol.
  • Further exemplary bioluminescent materials include, but are not limited to, luciferin and aequorin.
  • Further exemplary radioactive materials include, but are not limited to, Iodine-125, Carbon-14, Sulfur-35, Tritium and Phosphorus-32.
  • any additional agent described herein as well as the dosing schedule can depend on various parameters, including, but not limited to, the human patient's general health, and the administering physician's and/or human patient's discretion. Co-administration may be simultaneous or sequential. Any additional agent described herein, can be administered prior to (e.g., about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 12 hours, about 24 hours, about 48 hours, about 72 hours, about 96 hours, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, 8 weeks, or about 12 weeks before), concurrently with, or subsequent to (e.g., about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 12 hours, about 24 hours, about 48 hours, about 72 hours, about 96 hours, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks,
  • any agent described herein is administered about 1 minute apart, about 10 minutes apart, about 30 minutes apart, less than about 1 hour apart, about 1 hour apart, about 1 hour to about 2 hours apart, about 2 hours to about 3 hours apart, about 3 hours to about 4 hours apart, about 4 hours to about 5 hours apart, about 5 hours to about 6 hours apart, about 6 hours to about 7 hours apart, about 7 hours to about 8 hours apart, about 8 hours to about 9 hours apart, about 9 hours to about 10 hours apart, about 10 hours to about 11 hours apart, about 11 hours to about 12 hours apart, no more than about 24 hours apart or no more than about 48 hours apart.
  • the combination regimen is designed to exploit the finding that the nucleic acid drug dosing and formulation of the present invention has potent effects quickly (e.g. in about 6, or about 12, or about 24, or about 30, or about 36, or about 48 hours) and the effects can last about 7 days, or longer.
  • the dose of a nucleic acid drug is disclosed herein.
  • the dose of any additional agent that is useful is known to those in the art.
  • doses may be determined with reference Physicians' Desk Reference, 66th Edition, PDR Network; 2012 Edition (Dec. 27, 2011), the contents of which are incorporated by reference in its entirety.
  • the present invention allows a patient to receive doses that exceed those determined with reference Physicians' Desk Reference .
  • the dosage of any additional agent described herein can depend on several factors including the severity of the condition, whether the condition is to be treated or prevented, and the age, weight, and health of the human patient to be treated.
  • pharmacogenomic the effect of genotype on the pharmacokinetic, pharmacodynamic or efficacy profile of a therapeutic
  • dosage used may affect dosage used.
  • the exact individual dosages can be adjusted somewhat depending on a variety of factors, including the specific combination of the agents being administered, the time of administration, the route of administration, the nature of the formulation, the rate of excretion, the particular disease being treated, the severity of the disorder, and the anatomical location of the disorder. Some variations in the dosage can be expected.
  • nucleic acid delivery patch comprises a flexible membrane.
  • the nucleic acid delivery patch comprises a plurality of needles.
  • the plurality of needles are attached to the flexible membrane.
  • the patch comprises a nucleic acid.
  • the nucleic acid is present in solution.
  • the plurality of needles include one or more needles having a lumen.
  • the patch further comprises a second flexible membrane.
  • the flexible membrane and the second flexible membrane are arranged to form a cavity.
  • the cavity contains a nucleic acid.
  • the membrane comprises one or more holes through which a nucleic acid can pass.
  • one or more holes and one or more needles having a lumen are arranged to allow the passage of a solution containing a nucleic acid through at least one of the one or more holes and through at least one of the one or more needles having a lumen.
  • the patch is configured to deliver a solution to the skin.
  • the solution comprises a nucleic acid.
  • the solution comprises a vehicle.
  • the vehicle is a lipid or lipidoid.
  • the vehicle is a lipid-based transfection reagent.
  • the cell membrane can serve as a barrier to foreign nucleic acids. It has now been discovered that combining the patch of the present invention with an electric field can increase the efficiency of nucleic acid delivery. Certain embodiments are therefore directed to a nucleic acid delivery patch comprising a plurality of needles, wherein at least two needles form part of a high-voltage circuit. Certain embodiments are directed to an implantable “tattoo” for microneedle delivery (see, e.g. Nature Materials 12, pp 367-376 (2013), the contents of which are hereby incorporated by reference in their entirety). In one embodiment, the high-voltage circuit generates a voltage greater than about 10V. In another embodiment, the high-voltage circuit generates a voltage greater than about 20V.
  • an electric field is produced between two of the needles.
  • the magnitude of the electric field is at least about 100 V/cm.
  • the magnitude of the electric field is at least about 200 V/cm.
  • the patch is configured to deliver a nucleic acid to the epidermis.
  • the patch is configured to deliver a nucleic acid to the dermis.
  • the patch is configured to deliver a nucleic acid to sub-dermal tissue.
  • the patch is configured to deliver a nucleic acid to muscle.
  • Certain embodiments are directed to a nucleic acid delivery patch comprising a plurality of electrodes.
  • the plurality of electrodes is attached to a flexible membrane.
  • Other embodiments are directed to a nucleic acid delivery patch comprising a rigid structure. In one embodiment, a plurality of electrodes are attached to the rigid structure.
  • the compositions described herein are administered using an array of needles covering an affected area of the subject.
  • the treatment area is mechanically massaged after administration.
  • the treatment area is exposed to electric pulses after administration.
  • the electric pulses are between about 10V and about 200V for from about 50 microseconds to about 1 second.
  • the electric pulses are generated around the treatment area by a multielectrode array.
  • the present invention provides a patch delivery system, comprising a non-viral RNA transfection composition enclosed within a membrane, and an array of delivery needles delivering from about 10 ng to about 2000 ng of RNA per treatment area of about 100 cm 2 or less, or about 50 cm 2 or less, or about 10 cm 2 or less, or about 5 cm 2 or less, or about 1 cm 2 or less, or about 0.5 cm 2 or less, or about 0.2 cm 2 or less.
  • the non-viral transfection composition contains from about 10 ng to about 2000 ng per injection volume of about 20 ⁇ L to about 1 ml.
  • each needle delivers an injection volume of between 1 ⁇ L and 500 ⁇ L.
  • the delivery patch comprises an acrylic reservoir that holds the nucleic acid drug.
  • a silicon adhesive is added to create a semisolid suspension of microscopic, concentrated drug cells.
  • a patch that is associated with one or more enhancers include, without limitation, iontophoresis, ultrasound, chemicals including gels, microneedles, sonophoresis, lasers, and electroporatic methods).
  • the delivery is effected via a gel, optionally a hydro alcoholic gel containing a combination of enhancers (e.g. COMBIGEL (ANTARES PHARMA)).
  • a gel optionally a hydro alcoholic gel containing a combination of enhancers (e.g. COMBIGEL (ANTARES PHARMA)).
  • COMBIGEL ANTARES PHARMA
  • the RNA is delivered using needle arrays.
  • Illustrative needle arrays include, but are not limited to AdminPen 600 and those described in U.S. Pat. Nos. 7,658,728, 7,785,301, and 8,414,548, the entire disclosure of which are hereby incorporated by reference.
  • Other examples of needles include, for example, the 3MTM Hollow Microstructured Transdermal System and the 3M Solid Microstructured Transdermal Systems (sMTS). See, e.g. U.S. Pat. Nos. 3,034,507 and 3,675,766; Microneedles for Transdermal Drug Delivery. Advanced Drug Delivery Reviews. 56: 581-587 (2004); Pharm Res. 2011 Jan; 28(1): 31-40, the entire contents of which are hereby incorporated by reference in their entireties.
  • microneedles and/or microneedle arrays may be used.
  • the microneedles and/or microneedle arrays may be, without limitation, solid, RNA-coated, dissolving, biodegradable, and/or hollow.
  • the delivery is effected via a microneedle system, optionally combined with an electronically controlled micropump that delivers the drug at specific times or upon demand.
  • the MACROFLUX (Alza) system may be used.
  • inventions are directed to a method for delivering a nucleic acid to a cell in vivo comprising applying a nucleic acid to a tissue containing a cell in vivo.
  • the method further comprises applying a transient electric field in the vicinity of the cell.
  • the method results in the cell in vivo internalizing the nucleic acid.
  • the nucleic acid comprises synthetic RNA.
  • the method further results in the cell internalizing a therapeutically or cosmetically effective amount of the nucleic acid.
  • the cell is a skin cell.
  • the cell is a muscle cell.
  • the cell is a dermal fibroblast.
  • the cell is a keratinocyte. In a still further embodiment, the cell is a myoblast.
  • the nucleic acid comprises a protein of interest. In one embodiment, the protein of interest is a fluorescent protein. In another embodiment, the protein of interest is an extracellular-matrix protein.
  • the protein of interest is a member of the group: elastin, collagen, laminin, fibronectin, vitronectin, lysyl oxidase, elastin binding protein, a growth factor, fibroblast growth factor, transforming growth factor beta, granulocyte colony-stimulating factor, a matrix metalloproteinase, an actin, fibrillin, microfibril-associated glycoprotein, a lysyl-oxidase-like protein, platelet-derived growth factor, a lipase, an uncoupling protein, thermogenin, filaggrin, a fibroblast growth factor, an antibody, and a protein involved with pigment production.
  • elastin collagen, laminin, fibronectin, vitronectin, lysyl oxidase, elastin binding protein, a growth factor, fibroblast growth factor, transforming growth factor beta, granulocyte colony-stimulating factor, a matrix metalloproteinase, an
  • the method further comprises delivering the nucleic acid to the epidermis. In other embodiments, the method further comprises delivering the nucleic acid to the dermis. In still other embodiments, the method further comprises delivering the nucleic acid below the dermis. In one embodiment, the delivering is by injection. In another embodiment, the delivering is by injection using a micro-needle array. In yet another embodiment, the delivering is by topical administration. In a further embodiment, the delivering comprises disruption or removal of a part of the tissue. In a still further embodiment, the delivering comprises disruption or removal of the stratum corneum. In some embodiments, the nucleic acid is present in solution. In one embodiment, the solution comprises a growth factor.
  • the growth factor is a member of the group: a fibroblast growth factor and a transforming growth factor. In yet another embodiment, the growth factor is a member of the group: basis fibroblast growth factor and transforming growth factor beta. In other embodiments, the solution comprises cholesterol.

Abstract

The present invention relates in part to nucleic acids, including nucleic acids encoding proteins, therapeutics and cosmetics comprising nucleic acids, methods for delivering nucleic acids to cells, tissues, organs, and patients, methods for inducing cells to express proteins using nucleic acids, methods, kits and devices for transfecting, gene editing, and reprogramming cells, and cells, organisms, therapeutics, and cosmetics produced using these methods, kits, and devices.

Description

    PRIORITY
  • This application is a continuation of U.S. patent application Ser. No. 15/550,280, filed Aug. 10, 2017, which is a 371 National Stage application of PCT/US2016/018065, filed Feb. 1, 2016, which claims the benefit of and priority to U.S. Patent Application Nos. 62/116,232, filed Feb. 13, 2015, 62/195,462, filed Jul. 22, 2015, 62/242,383, filed Oct. 16, 2015, 62/245,726, filed Oct. 23, 2015; and 62/289,617, filed Feb. 1, 2016, the entire contents of all of which are incorporated by reference herein.
  • FIELD OF THE INVENTION
  • The present invention relates, in part, to methods, compositions, and products for producing and delivering nucleic acids to cells, tissues, organs, and patients, methods for expressing proteins in cells, tissues, organs, and patients, and cells, therapeutics, and cosmetics produced using these methods, compositions, and products.
  • DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY
  • The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (filename: FAB-009C1_ST25.txt; date recorded: Feb. 12, 2020; file size: 2,386,006 bytes).
  • BACKGROUND
  • Synthetic RNA and Nucleic-Acid Therapeutics
  • Ribonucleic acid (RNA) is ubiquitous in both prokaryotic and eukaryotic cells, where it encodes genetic information in the form of messenger RNA, binds and transports amino acids in the form of transfer RNA, assembles amino acids into proteins in the form of ribosomal RNA, and performs numerous other functions including gene expression regulation in the forms of microRNA and long non-coding RNA. RNA can be produced synthetically by methods including direct chemical synthesis and in vitro transcription, and can be administered to patients for therapeutic use. However, previously described synthetic RNA molecules are unstable and trigger a potent innate-immune response in human cells. In addition, methods for efficient non-viral delivery of nucleic acids to patients, organs, tissues, and cells in vivo have not been previously described. The many drawbacks of existing synthetic RNA technologies and methods for delivery of nucleic acids make them undesirable for therapeutic or cosmetic use.
  • Cell Reprogramming and Cell-Based Therapies
  • Cells can be reprogrammed by exposing them to specific extracellular cues and/or by ectopic expression of specific proteins, microRNAs, etc. While several reprogramming methods have been previously described, most that rely on ectopic expression require the introduction of exogenous DNA, which can carry mutation risks. DNA-free reprogramming methods based on direct delivery of reprogramming proteins have been reported. However, these methods are too inefficient and unreliable for commercial use. In addition, RNA-based reprogramming methods have been described (see, e.g., Angel. MIT Thesis. 2008. 1-56; Angel et al. PLoS ONE. 2010. 5, 107; Warren et al. Cell Stem Cell. 2010. 7, 618-630; Angel. MIT Thesis. 2011. 1-89; and Lee et al. Cell. 2012. 151, 547-558; the contents of all of which are hereby incorporated by reference). However, existing RNA-based reprogramming methods are slow, unreliable, and inefficient when performed on adult cells, require many transfections (resulting in significant expense and opportunity for error), can reprogram only a limited number of cell types, can reprogram cells to only a limited number of cell types, require the use of immunosuppressants, and require the use of multiple human-derived components, including blood-derived HSA and human fibroblast feeders. The many drawbacks of previously disclosed RNA-based reprogramming methods make them undesirable for research, therapeutic or cosmetic use.
  • Gene Editing Several naturally occurring proteins contain DNA-binding domains that can recognize specific DNA sequences, for example, zinc fingers (ZFs) and transcription activator-like effectors (TALEs). Fusion proteins containing one or more of these DNA-binding domains and the cleavage domain of FokI endonuclease can be used to create a double-strand break in a desired region of DNA in a cell (see, e.g., US Patent Appl. Pub. No. US 2012/0064620, US Patent Appl. Pub. No. US 2011/0239315, U.S. Pat. No. 8,470,973, US Patent Appl. Pub. No. US 2013/0217119, U.S. Pat. No. 8,420,782, US Patent Appl. Pub. No. US 2011/0301073, US Patent Appl. Pub. No. US 2011/0145940, U.S. Pat. Nos. 8,450,471, 8,440,431, 8,440,432, and US Patent Appl. Pub. No. 2013/0122581, the contents of all of which are hereby incorporated by reference). Other gene-editing proteins include clustered regularly interspaced short palindromic repeat (CRISPR)-associated proteins. However, current methods for gene editing cells are inefficient and carry a risk of uncontrolled mutagenesis, making them undesirable for research, therapeutic or cosmetic use. Methods for DNA-free gene editing of somatic cells have not been previously explored, nor have methods for simultaneous or sequential gene editing and reprogramming of somatic cells. In addition, methods for directly gene editing cells in patients (i.e., in vivo) have not been previously explored, and the development of such methods has been limited by a lack of acceptable targets, inefficient delivery, inefficient expression of the gene-editing protein/proteins, inefficient gene editing by the expressed gene-editing protein/proteins, due in part to poor binding of DNA-binding domains, excessive off-target effects, due in part to non-directed dimerization of the FokI cleavage domain and poor specificity of DNA-binding domains, and other factors. Finally, the use of gene editing in anti-bacterial, anti-viral, and anti-cancer treatments has not been previously explored.
  • Accordingly, there remains a need for improved methods and compositions for the production and delivery of nucleic acids to cells, tissues, organs, and patients.
  • SUMMARY OF THE INVENTION
  • The present invention provides, in part, compositions, methods, articles, and devices for delivering nucleic acids to cells, tissues, organs, and patients, methods for inducing cells to express proteins, methods, articles, and devices for producing these compositions, methods, articles, and devices, and compositions and articles, including cells, organisms, cosmetics and therapeutics, produced using these compositions, methods, articles, and devices. Unlike previously reported methods, certain embodiments of the present invention provide small doses of nucleic acids to achieve significant and lasting protein expression in humans.
  • In various aspects, the present invention is based on the surprising discovery of safe and effective doses and administration parameters for nucleic acid drugs in human subjects. In some aspects, there is provided a method for delivering a nucleic acid drug, comprising administering an effective dose of a nucleic acid drug to a human subject in need thereof. In various embodiments, the nucleic acid drug comprises a RNA comprising one or more non-canonical nucleotides (a/k/a “modified RNA”). In other embodiments, the effective dose is an amount sufficient to substantially increase an amount of a protein encoded by the nucleic acid drug in the human subject and/or substantially avoid an immune reaction in a human subject, wherein the immune reaction is optionally mediated by the innate immune system.
  • In some aspects, there is provided a method for expressing a protein of interest in a population of cells in a mammalian subject, comprising administering a non-viral transfection composition comprising an effective dose of a RNA encoding the protein of interest to said cells, where the transfection composition is administered in an amount that allows for expression of said protein in said cells for at least about six hours to about five days without substantial cellular toxicity. In some embodiments, the RNA contains one or more non-canonical nucleotides that avoid substantial cellular toxicity.
  • In some embodiments, the effective dose is about 100 ng to about 2000 ng (e.g., about, or no more than about, 100 ng, or 200 ng, or 300 ng, or 400 ng, or 500 ng, or 600 ng, or 700 ng, or 800 ng, or 900 ng, or 1000 ng, or 1100 ng, or 1200 ng, or 1300 ng, or 1400 ng, or 1500 ng, or 1600 ng, or 1700 ng, or 1800 ng, or 1900 ng, or 2000 ng). In other embodiments, the effective dose is less than about 100 ng. In certain embodiments, the effective dose is about 10 ng to about 100 ng (e.g., about, or no more than about, 10 ng, or 20 ng, or 30 ng, or 40 ng, or 50 ng, or 60 ng, or 70 ng, or 80 ng, or 90 ng, or 100 ng).
  • In some embodiments, the effective dose is about 1.4 ng/kg to about 30 ng/kg (e.g., about, or no more than about, 1.4 ng/kg, or 2.5 ng/kg, or 5 ng/kg, or 10 ng/kg, or 15 ng/kg, or 20 ng/kg, or 25 ng/kg, or 30 ng/kg. In other embodiments, the effective dose is less than about 1.5 ng/kg. In certain embodiments, the effective dose is about 0.14 ng/kg to about 1.4 ng/kg (e.g., about, or no more than about, 0.14 ng/kg, or 0.25 ng/kg, or 0.5 ng/kg, or 0.75 ng/kg, or 1 ng/kg, or 1.25 ng/kg, or 1.4 ng/kg).
  • In some embodiments, the effective dose is about 350 ng/cm2 to about 7000 ng/cm2 (e.g., about, or no more than about, 350 ng/cm2, or 500 ng/cm2, or 750 ng/cm2, or 1000 ng/cm2, or 2000 ng/cm2, or 3000 ng/cm2, or 4000 ng/cm2, or 5000 ng/cm2, or 6000 ng/cm2, or 7000 ng/cm2). In other embodiments, the effective dose is less than about 350 ng/cm2. In certain embodiments, the effective dose is about 35 ng/cm2 to about 350 ng/cm2 (e.g., about, or no more than about, 35 ng/cm2, or 50 ng/cm2, or 75 ng/cm2, or 100 ng/cm2, or 150 ng/cm2, or 200 ng/cm2, or 250 ng/cm2, or 300 ng/cm2, or 350 ng/cm2.
  • In some embodiments, the effective dose is about 0.28 picomoles to about 5.7 picomoles (e.g., about, or no more than about, 0.28 picomoles, or 0.5 picomoles, or 0.75 picomoles, or 1 picomole, or 2 picomoles, or 3 picomoles, or 4 picomoles, or 5 picomoles, or 5.7 picomoles). In other embodiments, the effective dose is less than about 0.28 picomoles. In certain embodiments, the effective dose is about 0.028 picomoles to about 0.28 picomoles (e.g., about, or no more than about, 0.028 picomoles, or 0.05 picomoles, or 0.075 picomoles, or 0.1 picomoles, or 0.15 picomoles, or 0.2 picomoles, or 0.25 picomoles, or 0.28 picomoles).
  • In some embodiments, the effective dose is about 0.004 picomoles/kg to about 0.082 picomoles/kg (e.g., about, or no more than about, 0.004 picomoles/kg, or 0.01 picomoles/kg, or 0.02 picomoles/kg, or 0.03 picomoles/kg, or 0.04 picomoles/kg, or 0.05 picomoles/kg, or 0.06 picomoles/kg, or 0.07 picomoles/kg, or 0.08 picomoles/kg, or 0.082 picomoles/kg). In other embodiments, the effective dose is less than about 0.004 picomoles/kg. In certain embodiments, the effective dose is about 0.0004 picomoles/kg to about 0.004 picomoles/kg (e.g., about, or no more than about, 0.0004 picomoles/kg, or 0.001 picomoles/kg, or 0.002 picomoles/kg, or 0.003 picomoles/kg, or 0.004 picomoles/kg).
  • In some embodiments, the effective dose is about 1 picomole/cm2 to about 20 picomoles/cm2 (e.g., about, or no more than about, 1 picomole/cm2, or 2 picomoles/cm2, or 3 picomoles/cm2, or 4 picomoles/cm2, or 5 picomoles/cm2, or 6 picomoles/cm2, or 7 picomoles/cm2, or 8 picomoles/cm2, or 9 picomoles/cm2, or 10 picomoles/cm2, or 12 picomoles/cm2, or 14 picomoles/cm2, or 16 picomoles/cm2, or 18 picomoles/cm2, or 20 picomoles/cm2). In other embodiments, the effective dose is less than about 1 picomole/cm2. In certain embodiments, the effective dose is about 0.1 picomoles/cm2 to about 1 picomole/cm2 (e.g., about, or no more than about, 0.1 picomoles/cm2, or 0.2 picomoles/cm2, or 0.3 picomoles/cm2, or 0.4 picomoles/cm2, or 0.5 picomoles/cm2, or 0.6 picomoles/cm2, or 0.7 picomoles/cm2, or 0.8 picomoles/cm2, or 0.9 picomoles/cm2, or 1 picomole/cm2).
  • In various embodiments, the nucleic acid drug is administered in a pharmaceutically acceptable formulation, including a formulation suitable for one or more of injection (e.g. subcutaneous injection, intradermal injection (including to the dermis or epidermis), subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection) and topical administration and/or for administration to the integumentary system (e.g. to one or more of the epidermis (optionally selected from the stratum corneum, stratum lucidum, stratum granulosum, stratum spinosum, and stratum germinativum), basement membrane, dermis (optionally selected from the papillary region and the reticular region), subcutis, and conjunctiva) and/or for administration to the eye (e.g., to one or more of the cornea, sclera, iris, lens, corneal limbus, optic nerve, choroid, ciliary body, anterior segment, anterior chamber, and retina).
  • In various embodiments, the nucleic acid drug is formulated with one or more lipids to enhance uptake of the nucleic acid drug by cells, the lipids optionally selected from Table 1. In other embodiments, the nucleic acid drug is formulated with one or more nanoparticles, optionally lipid or polymeric nanoparticles, to enhance uptake of the nucleic acid drug by cells, to enhance duration of protein expression, or to otherwise enhance the safety and/or efficacy of the nucleic acid drug.
  • In various embodiments, the nucleic acid drug is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose is administered to a surface area of about 4 mm2 to about 1000 mm2 (e.g. about, or no more than about, 4 mm2, or 5 mm2, or 10 mm2, or 25 mm2, or 50 mm2, or 75 mm2, or 100 mm2, or 125 mm2, or 150 mm2, or 200 mm2, or 500 mm2, or 1000 mm2).
  • In various embodiments, the nucleic acid drug is administered in a treatment regimen, optionally with an additional agent or adjuvant therapy described herein, and the administration is about weekly to about once every 24 weeks (e.g. about, or not more than about, weekly, or once every 2 weeks, or once every 3 weeks, or once every 4 weeks, or once every 5 weeks, or once every 6 weeks, or once every 7 weeks, or once every 8 weeks, or once every 9 weeks, or once every 9 weeks, or once every 9 weeks, or once every 9 weeks, or once every 10 weeks, or once every 11 weeks, or once every 12 weeks, or once every 13 weeks, or once every 14 weeks, or once every 15 weeks, or once every 20 weeks, or once every 24 weeks). In other embodiments, the nucleic acid drug is administered in a treatment regimen, optionally with an additional agent or adjuvant therapy described herein, and the administration is about daily to about weekly (e.g., about, or not more than about, daily, or once every 2 days, or once every 3 days, or once every 4 days, or once every 5 days, or once every 6 days, or weekly).
  • In various embodiments the nucleic acid drug comprises RNA comprising one or more non-canonical nucleotides, optionally having one or more substitutions at the 2C and/or 4C and/or 5C positions for a pyrimidine or the 6C and/or 7N and/or 8C positions for a purine. In various embodiments, the non-canonical nucleotide is one or more of the non-canonical nucleotides described herein, including, for example, 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methyluridine, 5-methylpseudouridine, 5-hydroxymethyluridine, 5-hydroxymethylpseudouridine, 5-carboxyuridine, 5-carboxypseudouridine, 5-formyluridine, 5-formylpseudouridine, 5-methoxyuridine, and 5-methoxypseudouridine. Further, the RNA comprising one or more non-canonical nucleotides may have one or more of a 5′-UTR comprising a Kozak consensus sequence, a 5′-UTR or 3′-UTR comprising a sequence that increases RNA stability in vivo (e.g. an alpha-globin or beta-globin 5′-UTR or an alpha-globin or beta-globin 3′-UTR), and a 3′ poly(A) tail from about 20 nucleotides to about 250 nucleotides in length (e.g. about 20, or about 30, or about 40, or about 50, or about 60, or about 70, or about 80, or about 90, or about 100, or about 110, or about 120, or about 130, or about 140, or about 150, or about 160, or about 170, or about 180, or about 190, or about 200, or about 210, or about 220, or about 230, or about 240, or about 250 nucleotides in length).
  • Further, some aspects of the methods described herein find use in various medical treatments, including, by way of illustration, treating a disease, disorder and/or condition of the integumentary system or altering, modifying and/or changing the integumentary system (e.g. cosmetically).
  • Also contemplated are kits suitable for use in human therapy, comprising the nucleic acid drug described herein in a unit dosage form of about 10 ng to about 2000 ng (e.g. about, or no more than about, 10 ng, or 20 ng, or 50 ng, or 100 ng, or 200 ng, or 300 ng, or 400 ng, or 500 ng, or 600 ng, or 700 ng, or 800 ng, or 900 ng, or 1000 ng, or 1100 ng, or 1200 ng, or 1300 ng, or 1400 ng, or 1500 ng, or 1600 ng, or 1700 ng, or 1800 ng, or 1900 ng, or 2000 ng) and an injection needle.
  • Further, in some aspects, the present invention provides a pharmaceutical formulation comprising the nucleic acid drug described herein and one or more of the vehicles (a/k/a “transfection reagents”, e.g., lipids) described herein, the formulation optionally being suitable for one or more of subcutaneous injection, intradermal injection, subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection, and topical administration.
  • In some aspects, nucleic acid delivery patches are provided. In one aspect, devices for delivering nucleic acids using electric fields are provided. Other aspects pertain to methods and compositions for delivery of nucleic acids to the skin. Still further aspects pertain to methods and compositions for expression of proteins in the skin.
  • In one aspect, the invention provides methods and compositions for treating diseases and conditions in humans, including, but not limited to, prophylactic treatments, treatments for rare diseases, including, but not limited to, dermatologic rare diseases, and treatments for use in medical dermatology and aesthetic medicine. In another aspect, the invention provides cosmetics comprising nucleic acids. Still further aspects relate to methods and compositions for altering pigmentation, for example, for the treatment of pigmentation disorders. Still further aspects relate to methods and compositions for enhancing healing, including, but not limited to, healing in response to a wound or surgery. The compositions of the present invention may alter, modify and/or change the appearance of a member of the integumenary system of a subject such as, but not limited, to skin, hair and nails. Such alteration, modification and/or change may be in the context of treatment methods and/or therapeutic uses as described herein including, by way of non-limiting example, dermatological treatments and cosmetics procedures.
  • Further, in various embodiments, the present invention relates to the targeting of various therapeutic proteins that are not limited to dermatological applications. For example, in various embodiments, the present compositions and methods find use in methods of treatment that are mediated by the increased expression of, for example, various soluble proteins as illustrated herein. In various embodiments, the nucleic acid drug encodes and/or increases the expression of one or more of a circulating protein, an extracellular matrix protein, an engineered protein, a gene-editing protein, a protein or peptide hormone, an enzyme, erythropoietin, darbepoetin alfa, NOVEPOETIN, elastin, collagen, an antibody or antibody fragment (e.g., a neutralizing antibody or antibody fragment), an intracellular protein, telomerase reverse transcriptase, a membrane protein, a fusion protein, a receptor, a ligand binding domain, a protein inhibitor, or a biologically active fragment, analogue or variant thereof. In other embodiments, administration of the nucleic acid drug results in one or more of an increase in hematocrit, an increase in tissue elasticity, an increase in tissue strength, and increase in skin hydration and/or water retention, hair growth, fat reduction, an insertion, deletion or mutation in DNA, conversion of a prodrug to an active drug, a decrease in tumor size and/or number, a decrease in plaque size and/or number, an increase in vascularization, a decrease in vascularization, an increase in visual acuity, a decrease in pain, an increase in cardiac output (e.g., ejection fraction and stroke volume), a decrease in abnormal heart rhythm, a decrease in fibrosis, a decrease in one or more adverse neurological symptoms, conditions, or disorders (e.g., depression, dysregulation of appetite, polyphagia, anorexia, dementia, headache, fatigue, numbness, tremors, and dizziness), a decrease in erectile dysfunction, an increase in vitality, an increase in pulmonary function, an increase in kidney function, an increase in liver function, an increase in insulin sensitivity, a decrease in insulin sensitivity, a decrease in inflammation, an increase in tear production, an improvement in hearing, an increase in auditory perception, a decrease in tinnitus, a reduction in perspiration, partial or total clearance of an infection, an increase in fertility, a decrease in fertility, inhibition or neutralization of a protein, recruitment or stimulation of one or more components of the immune system, lengthening of telomeres, inhibition of cellular senescence, an increase in replicative potential, reprogramming, proliferation, differentiation, and an increase in differentiation potential.
  • In some aspects, RNA molecules with low toxicity and high translation efficiency are provided. In one aspect, a cell-culture medium for high-efficiency in vivo transfection, reprogramming, and gene editing of cells is provided. Other aspects pertain to methods for producing RNA molecules encoding reprogramming proteins. Still further aspects pertain to methods for producing RNA molecules encoding gene-editing proteins.
  • In one aspect, the invention provides high-efficiency gene-editing proteins comprising engineered nuclease cleavage domains. In another aspect, the invention provides high-fidelity gene-editing proteins comprising engineered nuclease cleavage domains. Other aspects relate to high-efficiency gene-editing proteins comprising engineered DNA-binding domains. Still further aspects pertain to high-fidelity gene-editing proteins comprising engineered DNA-binding domains. Still further aspects relate to gene-editing proteins comprising engineered repeat sequences. Some aspects relate to methods for altering the DNA sequence of a cell by transfecting the cell with or inducing the cell to express a gene-editing protein. Other aspects relate to methods for altering the DNA sequence of a cell that is present in an in vitro culture. Still further aspects relate to methods for altering the DNA sequence of a cell that is present in vivo.
  • In some aspects, the invention provides methods for treating cancer comprising administering to a patient a therapeutically effective amount of a gene-editing protein or a nucleic-acid encoding a gene-editing protein. In one aspect, the gene-editing protein is capable of altering the DNA sequence of a cancer associated gene. In another aspect, the cancer-associated gene is the BIRC5 gene. Still other aspects relate to therapeutics comprising nucleic acids and/or cells and methods of using therapeutics comprising nucleic acids and/or cells for the treatment of, for example, type 1 diabetes, heart disease, including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, cystic fibrosis, sickle-cell anemia, thalassemia, Fanconi anemia, severe combined immunodeficiency, hereditary sensory neuropathy, xeroderma pigmentosum, Huntington's disease, muscular dystrophy, amyotrophic lateral sclerosis, Alzheimer's disease, cancer, and infectious diseases including hepatitis and HIV/AIDS. In some aspects, the nucleic acids comprise RNA. In other aspects, the RNA comprises one or more non-canonical nucleotides. In still other aspects, the nucleic acids are delivered to cells using a virus. In some aspects, the virus is a replication-competent virus. In other aspects, the virus is a replication-incompetent virus.
  • The details of the invention are set forth in the accompanying description below. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, illustrative methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
  • DETAILED DESCRIPTION OF THE FIGURES
  • FIG. 1 depicts primary adult human dermal fibroblasts transfected with RNA encoding green fluorescent protein (“GFP”) and comprising the indicated nucleotides.
  • FIG. 2 depicts the result of a gene-expression analysis of the primary adult human dermal fibroblasts of FIG. 1 using a one-step real-time RT-PCR and primers designed to amplify human interferon beta mRNA. Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 3 depicts the results of a gene-expression analysis of cells transfected with RNA comprising the indicated nucleotides, conducted as in FIG. 2. Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 4 depicts the results of a gene-expression analysis of cells transfected with RNA comprising the indicated nucleotides, conducted as in FIG. 2, and using primers designed to amplify the indicated transcripts. Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 5 depicts the results of a gene-expression analysis of primary human epidermal keratinocytes transfected with RNA encoding NOVEPOETIN, and comprising the indicated nucleotides, conducted as in FIG. 2. Data were normalized to the untransfected sample (“Neg.”). GAPDH was used as a loading control.
  • FIG. 6 depicts intradermal injection of a solution comprising RNA encoding GFP into the ventral forearm of a healthy, 33 year-old, 70 kg, male human subject.
  • FIG. 7 depicts a region of the ventral forearm of the subject shown in FIG. 6 after treatment with RNA comprising 5-methoxyuridine and encoding GFP (injection sites 1-3) or COL7 (injection site 4). The image was taken immediately following the final injection.
  • FIG. 8 depicts the region of FIG. 7, 24 hours after injection.
  • FIG. 9 depicts the results of fluorescent imaging of the region of FIG. 7, using the indicated fluorescent channels. The dose at each injection site is also indicated. Images were taken 24 hours after injection.
  • FIG. 10 depicts the results of fluorescent imaging of the region of FIG. 7, using the FITC fluorescent channel. The dose at each injection site is indicated. Images were taken 48 hours after injection.
  • FIG. 11 depicts the results of quantitative fluorescent imaging of the region of FIG. 7, using the FITC fluorescent channel. The horizontal axis indicates time after injection.
  • FIG. 12 depicts the results of fluorescent imaging of an independent experiment in which a region of the ventral forearm of as the subject shown in FIG. 6 treated with RNA comprising 5-methoxyuridine and encoding GFP. The image was taken 24 hours after injection.
  • FIG. 13 depicts the results of an ELISA designed to detect darbepoetin alfa in culture media of primary human epidermal keratinocytes transfected with RNA comprising the indicated nucleotides and encoding NOVEPOETIN.
  • FIG. 14 depicts the results of an ELISA designed to detect darbepoetin alfa in culture media of primary human epidermal keratinocytes transfected with RNA comprising the indicated nucleotides and encoding NOVEPOETIN.
  • FIG. 15 depicts the results of an ELISA designed to detect darbepoetin alfa in culture media of primary human epidermal keratinocytes transfected with RNA comprising the indicated nucleotides and encoding NOVEPOETIN.
  • FIG. 16 depicts primary human dermal fibroblasts transfected with RNA comprising 5-methoxyuridine and encoding hTERT. Cells were fixed and stained using an antibody targeting hTERT 24 hours after transfection.
  • FIG. 17 depicts primary adult human dermal fibroblasts transfected with RNA encoding green fluorescent protein (“GFP”), prepared and stored as indicated.
  • FIG. 18 depicts single administration of NOVECRIT induced a rapid increase and sustained level of NOVEPOIETIN in serum. The Y axis shows concentration of NOVEPOIETIN protein (mU/mL).
  • FIG. 19 depicts a single administration of NOVECRIT stimulated erythropoiesis, yielding elevated hematocrit for at least 14 days. The left panel shows % hematocrit on the Y axis, while the right panel shows % reticulocytes.
  • FIG. 20 depicts a table summarizing TNFα, IL-6, and IFNα cytokine levels in plasma samples collected from a maximum tolerated dose of NOVECRIT in male Sprague Dawley rats study of Example 35.
  • FIG. 21 depicts a SURVEYOR assay using the DNA of primary adult human dermal fibroblasts transfected with RNA TALENs targeting the sequence TGAGCAGAAGTGGCTCAGTG (SEQ ID NO:467) and TGGCTGTACAGCTACACCCC (SEQ ID NO: 468), located within the COL7A1 gene. The bands present in the +RNA lane indicate editing of a region of the gene that is frequently involved in dystrophic epidermolysis bullosa.
  • FIG. 22 depicts a SURVEYOR assay using the DNA of primary adult human dermal fibroblasts transfected with RNA TALENs targeting the sequence TTCCACTCCTGCAGGGCCCC (SEQ ID NO:469) and TCGCCCTTCAGCCCGCGTTC (SEQ ID NO:470), located within the COL7A1 gene. The bands present in the +RNA lane indicate editing of a region of the gene that is frequently involved in dystrophic epidermolysis bullosa.
  • FIG. 23 shows the immunogenicity of various synthetic RNA constructs in the context of a gene-editing (i.e. unmodified nucleotides “A,G,U,C”; pseudouridine only “psU”; 5-methylcytidine only “5mC”; both pseudouridine and 5-methylcytidine “psU+5mC”; and a negative control “neg”).
  • FIG. 24 shows the gene-editing activity in cells transfected with various synthetic RNA constructs (i.e. unmodified nucleotides “A,G,U,C”; psuedouridine only “psU”; 5-methylcytidine only “5mC”; both psuedouridine and 5-methylcytidine “psU+5mC”; and a negative control “neg”).
  • FIG. 25 depicts gene editing of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and a single-stranded DNA repair template (“RT”) of the indicated length. The presence of bands at the locations shown by asterisks (“*”) indicates successful gene editing.
  • FIG. 26 depicts gene editing of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and an 80 nt single-stranded DNA repair template (“RT”) at the indicated ratios of RNA to repair template. The presence of bands at the locations shown by asterisks (“*”) indicates successful gene editing.
  • FIG. 27 depicts gene correction of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and a single-stranded DNA repair template (“RT”) of the indicated length. The presence of bands at the locations shown by asterisks (“*”) indicates successful gene correction.
  • FIG. 28 depicts gene correction of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and an 80 nt single-stranded DNA repair template (“RT”) at the indicated ratios of RNA to repair template. The presence of bands at the locations shown by asterisks (“*”) indicates successful gene correction.
  • FIG. 29 depicts gene editing (“T7E1”) and correction (“Digestion”) of the COL7A1 gene in primary human epidermal keratinocytes transfected with RNA encoding TALENs and an 80 nt single-stranded DNA repair template (“RT”). The presence of bands at the locations shown by asterisks (“*”) indicates successful gene editing (“T7E1”) and correction (“Digestion”).
  • FIG. 30 depicts the amount of BMP7 protein secreted by primary human dermal fibroblasts and primary human epidermal keratinocytes transfected with RNA encoding the indicated BMP7 variant, measured by ELISA. Asterisks (“*”) indicate saturation of the ELISA assay.
  • FIG. 31 depicts the amount of parathyroid hormone secreted by primary human epidermal keratinocytes transfected with RNA encoding PTH, measured by ELISA.
  • FIG. 32 demonstrates that repeated administration of NOVECRIT stimulated erythropoiesis, yielding elevated hematocrit for at least 14 days. For each data set, the order of histograms from left to right is: Group 1, Group 2, Group 3, Group 4, and Group 5.
  • FIG. 33 provides an illustrative experimental design for studying the effects of RNAs encoding BMP7 variants for the prevention and treatment of diabetic nephropathy.
  • FIG. 34 depicts BMP7 protein levels in rats treated with RNAs encoding BMP7 variants as described in Example 42. Error bars indicate SEM (n=6).
  • FIG. 35 depicts the urine volume, urine albumin, and urine creatinine levels in rats treated with RNAs encoding BMP7 variants as described in Example 42. For each data set, the order of histograms from left to right is: urine volume, urine creatinine, and urine albumin.
  • FIG. 36 depicts the effect of RNAs encoding BMP7 variants in treating diabetic nephropathy as described in Example 42. For each data set, the order of histograms from left to right is: Group 6—vehicle and Group 7—FTB-2 (BMP7 variant A).
  • FIG. 37, panels A-I, depict the results of gene-expression analysis of human epidermal keratinocytes transfected with RNA encoding BDNF, BMP-2, BMP-6, IL-2, IL-6, IL-15, IL-22, LIF or FGF-21.
  • FIG. 38 depicts the results of gene-expression analysis of human epidermal keratinocytes transfected with RNA encoding IL-15 or IL-15 and IL-15RA.
  • FIG. 39 depicts the serum levels of FGF21, IL15, IL6, IL22, and Novepoietin following a single intradermal injection of various RNAs encoding these proteins as described in Example 45. Three rats were analyzed for each RNA tested.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based, in part, on the discovery of a safe and effective dosing strategy for nucleic acid drugs, including RNA, such as RNA comprising non-canonical (or “modified”) nucleotides, in humans. The inventors believe this to be the first report of safe and effective dosing of RNA molecules, including those comprising non-canonical nucleotides, in humans. Despite reports in the art that very large doses of RNA molecules are needed for mammalian dosing, and minimal therapeutic effect is achieved despite high dosing (see, e.g. US Patent Publication No. 2013/0245103), the present inventors have surprisingly managed to dose synthetic RNA in a human and achieve significant target protein expression with minimal immunological or other side effects.
  • In various embodiments, the present invention provides improved doses, formulations, administration and methods of use of nucleic acid drugs, which include RNA, which may contain non-canonical nucleotides (e.g. a residue other than adenine, guanine, thymine, uracil, and cytosine or the standard nucleoside, nucleotide, deoxynucleoside or deoxynucleotide derivatives thereof). In various embodiments, the RNA comprising non-canonical nucleotides leads to the expression of a protein encoded by the RNA, the protein often being one of therapeutic benefit (sometimes called the “target” or “protein of interest”). Further, this expression of therapeutic protein is achieved with minimal or negligible toxicity.
  • In various aspects, the present invention is based on the surprising discovery of safe and effective doses and administration parameters of nucleic acid drugs for human subjects. Nucleic acid drugs include a dsDNA molecule, a ssDNA molecule, a RNA molecule, a dsRNA molecule, a ssRNA molecule, a plasmid, an oligonucleotide, a synthetic RNA molecule, a miRNA molecule, an mRNA molecule, and an siRNA molecule. In various embodiments, the RNA comprises non-canonical nucleotides.
  • In some aspects, there is provided a method for delivering a nucleic acid drug, comprising administering an effective dose of a nucleic acid drug to a human subject in need thereof, wherein the nucleic acid drug comprises a synthetic RNA. In various embodiments, the effective dose is an amount sufficient to substantially increase an amount of a protein encoded by the nucleic acid drug in the human subject. For example, when the nucleic acid drug is a synthetic RNA comprising one or more modified nucleotides, the nucleic acid drug may result in higher protein expression than levels obtainable with a nucleic acid drug that does not comprise one or more modified nucleotides (e.g., RNA comprising the canonical nucleotides A, G, U, and C). In some embodiments, the nucleic acid drug results in about a 2-fold, or about a 3-fold, or about a 4-fold, or about a 5-fold, or about a 10-fold, or about a 15-fold, or about a 20-fold, or about a 25-fold, or about a 30-fold, or about a 35-fold, or about a 40-fold, or about a 45-fold, or about a 50-fold, or about a 100-fold increase in protein expression as compared to levels obtainable with a nucleic acid drug that does not comprise one or more modified nucleotides.
  • In some embodiments, the nucleic acid drug provides a sustained therapeutic effect that is optionally mediated by a sustained expression of target protein. For instance, in some embodiments, the therapeutic effect is present for over about 1 day, or over about 2 days, or over about 3 days, or over about 4 days, or over about 5 days, or over about 6 days, or over about 7 days, or over about 8 days, or over about 9 days, or over about 10 days, or over about 14 days after administration. In some embodiments, this sustained effect obviates the need for, or reduces the amount of, maintenance doses.
  • In some embodiments, the nucleic acid drug provides a sustained target protein level. For instance, in some embodiments, the target protein is present (e.g. in measurable amounts, e.g. in the serum of a patient to whom the nucleic acid drug has been administered) for over about 1 day, or over about 2 days, or over about 3 days, or over about 4 days, or over about 5 days, or over about 6 days, or over about 7 days, or over about 8 days, or over about 9 days, or over about 10 days, or over about 14 days after administration. In some embodiments, this sustained effect obviates the need for, or reduces the amount of, maintenance doses.
  • In various embodiments, the nucleic acid drug provides therapeutic action without sustained presence of the nucleic acid drug itself. In some embodiments, the nucleic acid drug is rapidly metabolized, for instance, within about 6 hours, or about 12 hours, or about 18 hours, or about 24 hours, or about 2 days, or about 3 days, or about 4 days, or about 5 days, or about 1 week from administration.
  • In various embodiments, the effective dose is an amount that substantially avoids cell toxicity in vivo. In various embodiments, the effective dose is an amount that substantially avoids an immune reaction in a human subject. For example, the immune reaction may be an immune response mediated by the innate immune system. Immune response can be monitored using markers known in the art (e.g. cytokines, interferons, TLRs). In some embodiments, the effective dose obviates the need for treatment of the human subject with immune suppressants agents (e.g. B18R) used to moderate the residual toxicity. Accordingly, in some embodiments, the present methods allow for dosing that provides increased protein expression and reduces toxicity.
  • In some embodiments, the immune response is reduced by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, about 99.9%, or greater than about 99.9% as compared to the immune response induced by a corresponding unmodified nucleic acid. In some embodiments, upregulation of one or more immune response markers is reduced by about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, about 99.9%, or greater than about 99.9% as compared to the upregulation of the one or more immune response markers induced by a corresponding unmodified nucleic acid. In some embodiments, the immune response marker comprises an mRNA or protein product of an interferon gene, including an interferon alpha gene, IFNB1, TLR3, RARRES3, EIF2AK2, STAT1, STAT2, IFIT1, IFIT2, IFIT3, IFIT5, OAS1, OAS2, OAS3, OASL, ISG20 or a fragment, variant, analogue, or family-member thereof. In some embodiments, the immune response marker comprises an mRNA or protein product of an TNF gene, including an TNF alpha gene, TNFRSF1A; TNFRSF1B; LTBR; TNFRSF4; CD40; FAS; TNFRSF6B; CD27; TNFRSF8; TNFRSF9; TNFRSF10A; TNFRSF10B; TNFRSF10C; TNFRSF10D; TNFRSF11A; TNFRSF11B; TNFRSF12A; TNFRSF13B; TNFRSF13C; TNFRSF14; NGFR; TNFRSF17; TNFRSF18; TNFRSF19; TNFRSF21; TNFRSF25; and EDA2R or a fragment, variant, analogue, or family-member thereof. In some embodiments, the immune response marker comprises an mRNA or protein product of an interleukin gene, including an IL-6 gene, IL-1; IL-2; IL-3; IL-4; IL-5; IL-6; IL-7; IL-8 or CXCL8; IL-9; IL-10; IL-11; IL-12; IL-13; IL-14; IL-15; IL-16; IL-17; IL-18; IL-19; IL-20; IL-21; IL-22; IL-23; IL-24; IL-25; IL-26; IL-27; IL-28; IL-29; IL-30; IL-31; IL-32; IL-33; IL-35; IL-36 or a fragment, variant, analogue, or family-member thereof.
  • In some embodiments, cell death is about 10%, about 25%, about 50%, about 75%, about 85%, about 90%, about 95%, or over about 95% less than the cell death observed with a corresponding unmodified nucleic acid. Moreover, cell death may affect fewer than about 50%, about 40%, about 30%, about 20%, about 10%, about 5%, about 1%, about 0.1%, about 0.01% or fewer than about 0.01% of cells contacted with the modified nucleic acids.
  • In some embodiments, there is provided a method for expressing a protein of interest in a population of cells in a mammalian subject, comprising administering a non-viral transfection composition comprising an effective dose of a RNA encoding the protein of interest to said cells, the RNA containing one or more non-canonical nucleotides that avoid substantial cellular toxicity, where the transfection composition is administered in an amount that allows for expression of said protein in said cells for at least about five days (e.g. about 5, or about 6, or about 7, about 8, or about 9, or about 10, or about 14 days) without substantial cellular toxicity. In some embodiments, there is provided a method for expressing a protein of interest in a population of cells in a mammalian subject, comprising administering a non-viral transfection composition comprising an effective dose of a RNA encoding the protein of interest to said cells, the RNA containing one or more non-canonical nucleotides that avoid substantial cellular toxicity, where the transfection composition is administered in an amount that allows for expression of said protein in said cells for at least about six hours (e.g. about six hours, or about 12 hours, or about 1 day, or about 2 days, or about 3 days, or about 4 days, or about 5 days) without substantial cellular toxicity.
  • In some embodiments, the effective dose of the nucleic acid drug, including synthetic RNA, is about 100 ng to about 2000 ng, or about 200 ng to about 1900 ng, or about 300 ng to about 1800 ng, or about 400 ng to about 1700 ng, or about 500 ng to about 1600 ng, or about 600 ng to about 1500 ng, or about 700 ng to about 1400 ng, or about 800 ng to about 1300 ng, or about 900 ng to about 1200 ng, or about 1000 ng to about 1100 ng, or about 500 ng to about 2000 ng, or about 500 ng to about 1500 ng, or about 500 ng to about 1000 ng, or about 1000 ng to about 1500 ng, or about 1000 ng to about 2000 ng, or about 1500 ng to about 2000 ng, or about 100 ng to about 500 ng, or about 200 ng to about 400 ng, or about 10 ng to about 100 ng, or about 20 ng to about 90 ng, or about 30 ng to about 80 ng, or about 40 ng to about 70 ng, or about 50 ng to about 60 ng.
  • In some embodiments, the effective dose of the nucleic acid drug, including synthetic RNA, is no more than about 50 ng, or about 100 ng, or about 200 ng, or about 300 ng, or about 400 ng, or about 500 ng, or about 600 ng, or about 700 ng, or about 800 ng, or about 900 ng, or about 1000 ng, or about 1100 ng, or about 1200 ng, or about 1300 ng, or about 1400 ng, or about 1500 ng, or about 1600 ng, or about 1700 ng, or about 1800 ng, or about 1900 ng, or about 2000 ng.
  • In some embodiments, the effective dose of the nucleic acid drug, including synthetic RNA, is about 50 ng, or about 100 ng, or about 200 ng, or about 300 ng, or about 400 ng, or about 500 ng, or about 600 ng, or about 700 ng, or about 800 ng, or about 900 ng, or about 1000 ng, or about 1100 ng, or about 1200 ng, or about 1300 ng, or about 1400 ng, or about 1500 ng, or about 1600 ng, or about 1700 ng, or about 1800 ng, or about 1900 ng, or about 2000 ng.
  • In some embodiments, the effective dose of the nucleic acid drug, including synthetic RNA, is about 0.028 pmol, or about 0.05 pmol, or about 0.1 pmol, or about 0.2 pmol, or about 0.3 pmol, or about 0.4 pmol, or about 0.5 pmol, or about 0.6 pmol, or about 0.7 pmol, or about 0.8 pmol, or about 0.9 pmol, or about 1.0 pmol, or about 1.2 pmol, or about 1.4 pmol, or about 1.6 pmol, or about 1.8 pmol, or about 2.0 pmol, or about 2.2 pmol, or about 2.4 pmol, or about 2.6 pmol, or about 2.8 pmol, or about 3.0 pmol, or about 3.2 pmol, or about 3.4 pmol, or about 3.6 pmol, or about 3.8 pmol, or about 4.0 pmol, or about 4.2 pmol, or about 4.4 pmol, or about 4.6 pmol, or about 4.8 pmol, or about 5.0 pmol, or about 5.5 pmol, or about 5.7 pmol.
  • In some embodiments, the nucleic acid drug, including synthetic RNA, is administered at a concentration of about 0.1 nM, or about 0.25 nM, or about 0.5 nM, or about 0.75 nM, or about 1 nM, or about 2.5 nM, or about 5 nM, or about 7.5 nM, or about 10 nM, or about 20 nM, or about 30 nM, or about 40 nM, or about 50 nM, or about 60 nM, or about 70 nM, or about 80 nM, or about 90 nM, or about 100 nM, or about 110 nM, or about 120 nM, or about 150 nM, or about 175 nM, or about 200 nM.
  • In some embodiments, the effective dose of the nucleic acid drug is about 350 ng/cm2, or about 500 ng/cm2, or about 750 ng/cm2, or about 1000 ng/cm2, or about 2000 ng/cm2, or about 3000 ng/cm2, or about 4000 ng/cm2, or about 5000 ng/cm2, or about 6000 ng/cm2, or about 7000 ng/cm2. In other embodiments, the effective dose is less than about 350 ng/cm2. In certain embodiments, the effective dose is about 35 ng/cm2, or about 50 ng/cm2, or about 75 ng/cm2, or about 100 ng/cm2, or about 150 ng/cm2, or about 200 ng/cm2, or about 250 ng/cm2, or about 300 ng/cm2, or about 350 ng/cm2.
  • In some embodiments, the effective dose of the nucleic acid drug is about 35 ng/cm2 to about 7000 ng/cm2, or about 50 ng/cm2 to about 5000 ng/cm2, or about 100 ng/cm2 to about 3000 ng/cm2, or about 500 ng/cm2 to about 2000 ng/cm2, or about 750 ng/cm2 to about 1500 ng/cm2, or about 800 ng/cm2 to about 1200 ng/cm2, or about 900 ng/cm2 to about 1100 ng/cm2.
  • In some embodiments, the effective dose of the nucleic acid drug is about 1 picomole/cm2, or about 2 picomoles/cm2, or about 3 picomoles/cm2, or about 4 picomoles/cm2, or about 5 picomoles/cm2, or about 6 picomoles/cm2, or about 7 picomoles/cm2, or about 8 picomoles/cm2, or about 9 picomoles/cm2, or about 10 picomoles/cm2, or about 12 picomoles/cm2, or about 14 picomoles/cm2, or about 16 picomoles/cm2, or about 18 picomoles/cm2, or about 20 picomoles/cm2. In other embodiments, the effective dose is less than about 1 picomole/cm2. In certain embodiments, the effective dose is about 0.1 picomoles/cm2, or about 0.2 picomoles/cm2, or about 0.3 picomoles/cm2, or about 0.4 picomoles/cm2, or about 0.5 picomoles/cm2, or about 0.6 picomoles/cm2, or about 0.7 picomoles/cm2, or about 0.8 picomoles/cm2, or about 0.9 picomoles/cm2, or about 1 picomole/cm2.
  • In some embodiments, the effective dose of the nucleic acid drug is about 0.1 picomoles/cm2 to about 20 picomoles/cm2, or about 0.2 picomoles/cm2 to about 15 picomoles/cm2, or about 0.5 picomoles/cm2 to about 10 picomoles/cm2, or about 0.8 picomoles/cm2 to about 8 picomoles/cm2, or about 1 picomole/cm2 to about 5 picomoles/cm2, or about 2 picomoles/cm2 to about 4 picomoles/cm2.
  • In various embodiments, the nucleic acid drug, including synthetic RNA, is administered in a pharmaceutically acceptable formulation. In various embodiments, the nucleic acid drug, including synthetic RNA, is formulated for one or more of injection and topical administration. By way of example, the nucleic acid drug, including synthetic RNA, may be formulated for injection to a tissue of interest, e.g. a disease site (by way of non-limiting example, a tumor). In various embodiments, injection includes delivery via a patch. In some embodiments, the delivery is mediated by electrical stimulation. In various embodiments, the nucleic acid drug, including synthetic RNA, is formulated for administration to one or more of the epidermis (optionally selected from the stratum corneum, stratum lucidum, stratum granulosum, stratum spinosum, and stratum germinativum), basement membrane, dermis (optionally selected from the papillary region and the reticular region), subcutis, conjunctiva cornea, sclera, iris, lens, corneal limbus, optic nerve, choroid, ciliary body, anterior segment, anterior chamber, and retina. In various embodiments, the nucleic acid drug, including synthetic RNA, is formulated for one or more of subcutaneous injection, intradermal injection, subdermal injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, intratumoral injection, and topical administration. In various embodiments, the nucleic acid drug, including synthetic RNA, is formulated for intradermal (ID) injection to one or more of the dermis or epidermis. In various embodiments, the nucleic acid drug, including synthetic RNA, is administered in a manner such that it effects one or more of keratinocytes and fibroblasts (e.g. causes these cells to express one or more therapeutic proteins).
  • Accordingly, the present invention provides various formulations as described herein. Further, in some embodiments, the formulations described herein find use in the various delivery and/or treatment methods of the present invention. For instance, formulations can comprise a vesicle, for instance, a liposome (see Langer, 1990, Science 249:1527-1533; Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989). In various embodiments, the formulation comprises an aqueous suspension of liposomes. Illustrative liposome components are set forth in Table 1, and are given by way of example, and not by way of limitation. In various embodiments, one or more, or two or more, or three or more, or four or more, or five or more of the lipids of Table 1 are combined in a formulation.
  • TABLE 1
    Illustrative Biocompatible Lipids
    1 3β-[N-(N′,N′-dimethylaminoethane)-carbamoyl]cholesterol
    (DC-Cholesterol)
    2 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP/18:1 TAP)
    3 N-(4-carboxybenzyl)-N,N-dimethyl-2,3-bis(oleoyloxy)propan-
    1-aminium (DOBAQ)
    4 1,2-dimyristoyl-3-trimethylammonium-propane (14:0 TAP)
    5 1,2-dipalmitoyl-3-trimethylammonium-propane (16:0 TAP)
    6 1,2-stearoyl-3-trimethylammonium-propane (18:0 TAP)
    7 1,2-dioleoyl-3-dimethylammonium-propane (DODAP/18:1 DAP)
    8 1,2-dimyristoyl-3-dimethylammonium-propane (14:0 DAP)
    9 1,2-dipalmitoyl-3-dimethylammonium-propane (16:0 DAP)
    10 1,2-distearoyl-3-dimethylammonium-propane (18:0 DAP)
    11 dimethyldioctadecylammonium (18:0 DDAB)
    12 1,2-dilauroyl-sn-glycero-3-ethylphosphocholine (12:0 EthylPC)
    13 1,2-dimyristoyl-sn-glycero-3-ethylphosphocholine (14:0 EthylPC)
    14 1,2-dimyristoleoyl-sn-glycero-3-ethylphosphocholine (14:1 EthylPC)
    15 1,2-dipalmitoyl-sn-glycero-3-ethylphosphocholine (16:0 EthylPC)
    16 1,2-distearoyl-sn-glycero-3-ethylphosphocholine (18:0 EthylPC)
    17 1,2-dioleoyl-sn-glycero-3-ethylphosphocholine (18:1 EthylPC)
    18 1-palmitoyl-2-oleoyl-sn-glycero-3-ethylphosphocholine
    (16:1-18:1 EthylPC)
    19 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA)
    20 N1-[2-((1S)-1-[(3-aminopropyl)amino]-4-[di(3-amino-
    propyl)amino]butylcarboxamido)ethyl]-3,4-di[oleyloxy]-benzamide
    (MVL5)
    21 2,3-dioleyloxy-N-[2-spermine carboxamide]ethyl-N,N-dimethyl-1-
    propanammonium trifluoroacetate (DOSPA)
    22 1,3-di-oleoyloxy-2-(6-carboxy-spermyl)-propylamid (DOSPER)
    23 N-[1-(2,3-dimyristyloxy)propyl]-N,N-dimethyl-N-(2-
    hydroxyethyl)ammonium bromide (DMRIE)
    24 LIPOFECTAMINE, LIPOFECTAMINE 2000, LIPOFECTAMINE
    RNAiMAX, LIPOFECTAMINE 3000, LIPOFECTAMINE
    MessengerMAX, TransIT mRNA
    25 dioctadecyl amidoglyceryl spermine (DOGS)
    26 dioleoyl phosphatidyl ethanolamine (DOPE)
  • In some embodiments, the liposomes include LIPOFECTAMINE 3000. In some embodiments, the liposomes include one or more lipids described in U.S. Pat. No. 4,897,355 or 7,479,573 or in International Patent Publication No. WO/2015/089487, or in Feigner, P. L. et al. (1987) Proc. Natl. Acad. Sci. USA 84:7413-7417, the entire contents of each is incorporated by reference in their entireties).
  • In some embodiments, the liposome comprises N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA). In some embodiments, the liposome comprises dioleoylphosphatidylethanolamine (DOPE).
  • In one embodiment, the liposomes include one or more polyethylene glycol (PEG) chains, optionally selected from PEG200, PEG300, PEG400, PEG600, PEG800, PEG1000, PEG1500, PEG2000, PEG3000, and PEG4000. In some embodiments, the PEG is PEG2000. In some embodiments, the liposomes include 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) or a derivative thereof.
  • In some embodiments, the formulation comprises one or more of N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-DSPE), fully hydrogenated phosphatidylcholine, cholesterol, LIPOFECTAMINE 3000, a cationic lipid, a polycationic lipid, and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[folate(polyethylene glycol)-5000] (FA-MPEG5000-DSPE).
  • In one embodiment, the formulation comprises about 3.2 mg/mL N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-DSPE), about 9.6 mg/mL fully hydrogenated phosphatidylcholine, about 3.2 mg/mL cholesterol, about 2 mg/mL ammonium sulfate, and histidine as a buffer, with about 0.27 mg/mL 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[folate(polyethylene glycol)-5000](FA-MPEG5000-DSPE) added to the lipid mixture. In another embodiment, the nucleic acids are complexed by combining 1 μL of LIPOFECTAMINE 3000 per about 1 μg of nucleic acid and incubating at room temperature for at least about 5 minutes. In one embodiment, the LIPOFECTAMINE 3000 is a solution comprising a lipid at a concentration of about 1 mg/mL. In some embodiments, nucleic acids are encapsulated by combining about 10 μg of the liposome formulation per about 1 μg of nucleic acid and incubating at room temperature for about 5 minutes.
  • In some embodiments, the formulation comprises one or more nanoparticles. In one embodiment, the nanoparticle is a polymeric nanoparticle. In various embodiments, the formulation comprises one or more of a diblock copolymer, a triblock copolymer, a tetrablock copolymer, and a multiblock copolymer. In various embodiments, the formulation comprises one or more of polymeric nanoparticles comprising a polyethylene glycol (PEG)-modified polylactic acid (PLA) diblock copolymer (PLA-PEG) and PEG-polypropylene glycol-PEG-modified PLA-tetrablock copolymer (PLA-PEG-PPG-PEG).
  • In some embodiments, the formulation comprises one or more lipids that are described in WO/2000/027795, the entire contents of which are hereby incorporated by reference.
  • In one embodiment, the therapeutic comprises one or more ligands. In another embodiment, the therapeutic comprises at least one of: androgen, CD30 (TNFRSF8), a cell-penetrating peptide, CXCR, estrogen, epidermal growth factor, EGFR, HER2, folate, insulin, insulin-like growth factor-I, interleukin-13, integrin, progesterone, stromal-derived-factor-1, thrombin, vitamin D, and transferrin or a biologically active fragment or variant thereof.
  • The active compositions of the present invention may include classic pharmaceutical preparations. Administration of these compositions according to the present invention may be via any common route so long as the target tissue is available via that route. This includes oral, nasal, or buccal. Alternatively, administration may be by intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection, or by direct injection into diseased, e.g. cancer, tissue. The agents disclosed herein may also be administered by catheter systems. Such compositions would normally be administered as pharmaceutically acceptable compositions as described herein.
  • Administration of the compositions described herein may be, for example, by injection, topical administration, ophthalmic administration and intranasal administration. The injection, in some embodiments, may be linked to an electrical force (e.g. electroporation, including with devices that find use in electrochemotherapy (e.g. CLINIPORATOR, IGEA Srl, Carpi [MO], Italy)). The topical administration may be, but is not limited to, a cream, lotion, ointment, gel, spray, solution and the like. The topical administration may further include a penetration enhancer such as, but not limited to, surfactants, fatty acids, bile salts, chelating agents, non-chelating non-surfactants, polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether, fatty acids and/or salts in combination with bile acids and/or salts, sodium salt in combination with lauric acid, capric acid and UDCA, and the like. The topical administration may also include a fragrance, a colorant, a sunscreen, an antibacterial and/or a moisturizer. The compositions described herein may be administered to at least one site such as, but not limited to, forehead, scalp, hair follicles, hair, upper eyelids, lower eyelids, eyebrows, eyelashes, infraorbital area, periorbital areas, temple, nose, nose bridge, cheeks, tongue, nasolabial folds, lips, periobicular areas, jaw line, ears, neck, breast, forearm, upper arm, palm, hand, finger, nails, back, abdomen, sides, buttocks, thigh, calf, feet, toes and the like.
  • Routes of administration include, for example: intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, or topically, particularly to the ears, nose, eyes, or skin. In some embodiments, the administering is effected orally or by parenteral injection.
  • Upon formulation, solutions may be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective, as described herein. The formulations may easily be administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like. For parenteral administration in an aqueous solution, for example, the solution generally is suitably buffered and the liquid diluent first rendered isotonic with, for example, sufficient saline or glucose. Such aqueous solutions may be used, for example, for intravenous, intramuscular, subcutaneous and intraperitoneal administration. Preferably, sterile aqueous media are employed as is known to those of skill in the art, particularly in light of the present disclosure.
  • In various embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose is administered to a surface area of about 4 mm2 to about 150 mm2 (e.g. about, or no more than about, 4 mm2, or about 5 mm2, or about 6 mm2, or about 7 mm2, or about 8 mm2, or about 10 mm2, or about 20 mm2, or about 50 mm2, or about 100 mm2, or about 150 mm2). In various embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose administered to a surface area of no more than about 4 mm2, or about 5 mm2, or about 6 mm2, or about 7 mm2, or about 8 mm2, or about 10 mm2, or about 20 mm2, or about 50 mm2, or about 100 mm2, or about 150 mm2. In various embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose administered to a surface area of about 4 mm2, or about 5 mm2, or about 6 mm2, or about 7 mm2, or about 8 mm2, or about 10 mm2, or about 20 mm2, or about 50 mm2, or about 100 mm2, or about 150 mm2.
  • In various embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose (weight RNA/surface area of injection) is about 35 ng/cm2 to about 7000 ng/cm2. In various embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose (weight RNA/surface area of injection) is no more than about 35 ng/cm2, or about 50 ng/cm2, or about 75 ng/cm2, or about 100 ng/cm2, or about 125 ng/cm2, or about 150 ng/cm2, or about 175 ng/cm2, or about 200 ng/cm2, or about 225 ng/cm2, or about 250 ng/cm2, or about 500 ng/cm2, or about 1000 ng/cm2, or about 2000 ng/cm2, or about 5000 ng/cm2, or about 7000 ng/cm2. In various embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, and/or formulations comprising the same, is administered locally, optionally by one or more of subcutaneous injection, intradermal injection, subdermal injection and intramuscular injection, and the effective dose (weight RNA/surface area of injection) is about 35 ng/cm2, or about 50 ng/cm2, or about 75 ng/cm2, or about 100 ng/cm2, or about 125 ng/cm2, or about 150 ng/cm2, or about 175 ng/cm2, or about 200 ng/cm2, or about 225 ng/cm2, or about 250 ng/cm2, or about 500 ng/cm2, or about 1000 ng/cm2, or about 2000 ng/cm2, or about 5000 ng/cm2, or about 7000 ng/cm2.
  • Pharmaceutical preparations may additionally comprise delivery reagents (a.k.a. “transfection reagents”, a.k.a. “vehicles”, a.k.a. “delivery vehicles”) and/or excipients. Pharmaceutically acceptable delivery reagents, excipients, and methods of preparation and use thereof, including methods for preparing and administering pharmaceutical preparations to patients (a.k.a. “subjects”) are well known in the art, and are set forth in numerous publications, including, for example, in US Patent Appl. Pub. No. US 2008/0213377, the entirety of which is incorporated herein by reference.
  • For example, the present compositions can be in the form of pharmaceutically acceptable salts. Such salts include those listed in, for example, J. Pharma. Sci. 66, 2-19 (1977) and The Handbook of Pharmaceutical Salts; Properties, Selection, and Use. P. H. Stahl and C. G. Wermuth (eds.), Verlag, Zurich (Switzerland) 2002, which are hereby incorporated by reference in their entirety. Non-limiting examples of pharmaceutically acceptable salts include: sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, pamoate, phenylacetate, trifluoroacetate, acrylate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, o-acetoxybenzoate, naphthalene-2-benzoate, isobutyrate, phenylbutyrate, α-hydroxybutyrate, butyne-1,4-dicarboxylate, hexyne-1,4-dicarboxylate, caprate, caprylate, cinnamate, glycollate, heptanoate, hippurate, malate, hydroxymaleate, malonate, mandelate, mesylate, nicotinate, phthalate, teraphthalate, propiolate, propionate, phenylpropionate, sebacate, suberate, p-bromobenzenesulfonate, chlorobenzenesulfonate, ethylsulfonate, 2-hydroxyethylsulfonate, methylsulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, naphthalene-1,5-sulfonate, xylenesulfonate, tartarate salts, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N-methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-lower alkylamines), such as mono-; bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxyl-lower alkyl)-amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2-hydroxyethyl)amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like.
  • The present pharmaceutical compositions can comprises excipients, including liquids such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The pharmaceutical excipients can be, for example, saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like. In addition, auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used. In one embodiment, the pharmaceutically acceptable excipients are sterile when administered to a subject. Suitable pharmaceutical excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Any agent described herein, if desired, can also comprise minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Dosage forms suitable for parenteral administration (e.g. subcutaneous, intradermal, subdermal, intramuscular, intravenous, intraperitoneal, intra-articular, and infusion) include, for example, solutions, suspensions, dispersions, emulsions, and the like. They may also be manufactured in the form of sterile solid compositions (e.g. lyophilized composition), which can be dissolved or suspended in sterile injectable medium immediately before use. They may contain, for example, suspending or dispersing agents known in the art.
  • In some embodiments, the formulations described herein may comprise albumin and a nucleic acid molecule.
  • In some embodiments, the invention relates to a cosmetic composition. In one embodiment, the cosmetic composition comprises albumin. In another embodiment, the albumin is treated with an ion-exchange resin or charcoal. In yet another embodiment, the cosmetic composition comprises a nucleic acid molecule. In a further embodiment, the cosmetic composition comprises both albumin and a nucleic acid molecule. Still other embodiments are directed to a cosmetic treatment article comprising a cosmetic composition contained in a device configured to deliver the composition to a patient. Still other embodiments are directed to a device configured to deliver a cosmetic composition to a patient. In one embodiment, the nucleic acid molecule encodes a member of the group: elastin, collagen, tyrosinase, melanocortin 1 receptor, keratin, filaggren, an antibody, and hyaluronan synthase or a biologically active fragment, variant, analogue or family member thereof.
  • In some embodiments, the present invention provides treatment regimens. The inventors have discovered that the doses and administration described herein can produce a substantial protein expression effect quickly (e.g. in about 6, or about 12, or about 24, or about 36, or about 48 hours). Further, these effects can be sustained for about 7 days, or longer. In some embodiments, the present methods provide for administration of a nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, about weekly to about once every 20 weeks.
  • In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered about weekly, for at least 2 weeks (e.g. 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10 weeks). In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered about every other week for at least one month (e.g. 1, or 2, or 3, or 4, or 5, or 6, or 12 months). In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered monthly or about every other month. In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered is administered for at least two months, or at least 4 months, or at least 6 months, or at least 9 months, or at least one year.
  • In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered about weekly, or about once every 2 weeks, or about once every 3 weeks, or about once every 4 weeks, or about once every 5 weeks, or about once every 6 weeks, or about once every 7 weeks, or about once every 8 weeks, or about once every 9 weeks, or about once every 10 weeks, or about once every 11 weeks, or about once every 12 weeks, or about once every 13 weeks, or about once every 14 weeks, or about once every 15 weeks, or about once every 20 weeks, or about once every 24 weeks.
  • In some embodiments, the nucleic acid drug, including RNA comprising one or more non-canonical nucleotides, is administered no more than about weekly, or about once every 2 weeks, or about once every 3 weeks, or about once every 4 weeks, or about once every 5 weeks, or about once every 6 weeks, or about once every 7 weeks, or about once every 8 weeks, or about once every 9 weeks, or about once every 10 weeks, or about once every 11 weeks, or about once every 12 weeks, or about once every 13 weeks, or about once every 14 weeks, or about once every 15 weeks, or about once every 20 weeks, or about 24 weeks.
  • Certain proteins have long half-lives, and can persist in tissues for several hours, days, weeks, months or years. It has now been discovered that certain methods of treating a patient can result in accumulation of one or more proteins, including, for example, one or more beneficial proteins. Certain embodiments are therefore directed to a method for treating a patient comprising delivering to a patient in a series of doses a nucleic acid encoding one or more proteins. In one embodiment the nucleic acid comprises RNA comprising one or more non-canonical nucleotides. In another embodiment, a first dose is given at a first time-point. In yet another embodiment, a second dose is given at a second time-point. In a further embodiment, the amount of at least one of the one or more proteins in the patient at the second time-point is greater than the amount of said protein at the first time-point. In a still further embodiment, the method results in the accumulation of said protein in the patient.
  • In various embodiments, the present invention relates to nucleic acid drugs, which, in various embodiments are RNA comprising one or more non-canonical nucleotides. Certain non-canonical nucleotides, when incorporated into RNA molecules, can reduce the toxicity of the RNA molecules, in part, without wishing to be bound by theory, by interfering with binding of proteins that detect exogenous nucleic acids, for example, protein kinase R, Rig-1 and the oligoadenylate synthetase family of proteins. Non-canonical nucleotides that have been reported to reduce the toxicity of RNA molecules when incorporated therein include: pseudouridine, 5-methyluridine, 2-thiouridine, 5-methylcytidine, N6-methyladenosine, and certain combinations thereof. However, the chemical characteristics of non-canonical nucleotides that can enable them to lower the in vivo toxicity of RNA molecules have, until this point, remained unknown. Furthermore, incorporation of large amounts of most non-canonical nucleotides, for example, 5-methyluridine, 2-thiouridine, 5-methylcytidine, and N6-methyladenosine, can reduce the efficiency with which RNA molecules can be translated into protein, limiting the utility of RNA molecules containing these nucleotides in applications that require protein expression. In addition, while pseudouridine can be completely substituted for uridine in RNA molecules without reducing the efficiency with which the synthetic RNA molecules can be translated into protein, in certain situations, for example, when performing frequent, repeated transfections, synthetic RNA molecules containing only adenosine, guanosine, cytidine, and pseudouridine can exhibit excessive toxicity.
  • It has now been discovered that, and in some embodiments the invention pertains to, RNA molecules containing one or more non-canonical nucleotides that include one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine can be less toxic than synthetic RNA molecules containing only canonical nucleotides, due in part to the ability of substitutions at these positions to interfere with recognition of synthetic RNA molecules by proteins that detect exogenous nucleic acids, and furthermore, that substitutions at these positions can have minimal impact on the efficiency with which the synthetic RNA molecules can be translated into protein, due in part to the lack of interference of substitutions at these positions with base-pairing and base-stacking interactions.
  • Figure US20200261599A1-20200820-C00001
  • Examples of non-canonical nucleotides that include one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine include, but are not limited to: 2-thiouridine, 5-azauridine, pseudouridine, 4-thiouridine, 5-methyluridine, 5-methylpseudouridine, 5-aminouridine, 5-aminopseudouridine, 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methoxyuridine, 5-methoxypseudouridine, 5-hydroxymethyluridine, 5-hydroxymethylpseudouridine, 5-carboxyuridine, 5-carboxypseudouridine, 5-formyluridine, 5-formylpseudouridine, 5-methyl-5-azauridine, 5-amino-5-azauridine, 5-hydroxy-5-azauridine, 5-methylpseudouridine, 5-aminopseudouridine, 5-hydroxypseudouridine, 4-thio-5-azauridine, 4-thiopseudouridine, 4-thio-5-methyluridine, 4-thio-5-aminouridine, 4-thio-5-hydroxyuridine, 4-thio-5-methyl-5-azauridine, 4-thio-5-amino-5-azauridine, 4-thio-5-hydroxy-5-azauridine, 4-thio-5-methylpseudouridine, 4-thio-5-aminopseudouridine, 4-thio-5-hydroxypseudouridine, 2-thiocytidine, 5-azacytidine, pseudoisocytidine, N4-methylcytidine, N4-aminocytidine, N4-hydroxycytidine, 5-methylcytidine, 5-aminocytidine, 5-hydroxycytidine, 5-methoxycytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytydine, 5-methyl-5-azacytidine, 5-amino-5-azacytidine, 5-hydroxy-5-azacytidine, 5-methylpseudoisocytidine, 5-aminopseudoisocytidine, 5-hydroxypseudoisocytidine, N4-methyl-5-azacytidine, N4-methylpseudoisocytidine, 2-thio-5-azacytidine, 2-thiopseudoisocytidine, 2-thio-N4-methylcytidine, 2-thio-N4-aminocytidine, 2-thio-N4-hydroxycytidine, 2-thio-5-methylcytidine, 2-thio-5-aminocytidine, 2-thio-5-hydroxycytidine, 2-thio-5-methyl-5-azacytidine, 2-thio-5-amino-5-azacytidine, 2-thio-5-hydroxy-5-azacytidine, 2-thio-5-methylpseudoisocytidine, 2-thio-5-aminopseudoisocytidine, 2-thio-5-hydroxypseudoisocytidine, 2-thio-N4-methyl-5-azacytidine, 2-thio-N4-methylpseudoisocytidine, N4-methyl-5-methylcytidine, N4-methyl-5-aminocytidine, N4-methyl-5-hydroxycytidine, N4-methyl-5-methyl-5-azacytidine, N4-methyl-5-amino-5-azacytidine, N4-methyl-5-hydroxy-5-azacytidine, N4-methyl-5-methylpseudoisocytidine, N4-methyl-5-aminopseudoisocytidine, N4-methyl-5-hydroxypseudoisocytidine, N4-amino-5-azacytidine, N4-aminopseudoisocytidine, N4-amino-5-methylcytidine, N4-amino-5-aminocytidine, N4-amino-5-hydroxycytidine, N4-amino-5-methyl-5-azacytidine, N4-amino-5-amino-5-azacytidine, N4-amino-5-hydroxy-5-azacytidine, N4-amino-5-methylpseudoisocytidine, N4-amino-5-aminopseudoisocytidine, N4-amino-5-hydroxypseudoisocytidine, N4-hydroxy-5-azacytidine, N4-hydroxypseudoisocytidine, N4-hydroxy-5-methylcytidine, N4-hydroxy-5-aminocytidine, N4-hydroxy-5-hydroxycytidine, N4-hydroxy-5-methyl-5-azacytidine, N4-hydroxy-5-amino-5-azacytidine, N4-hydroxy-5-hydroxy-5-azacytidine, N4-hydroxy-5-methylpseudoisocytidine, N4-hydroxy-5-aminopseudoisocytidine, N4-hydroxy-5-hydroxypseudoisocytidine, 2-thio-N4-methyl-5-methylcytidine, 2-thio-N4-methyl-5-aminocytidine, 2-thio-N4-methyl-5-hydroxycytidine, 2-thio-N4-methyl-5-methyl-5-azacytidine, 2-thio-N4-methyl-5-amino-5-azacytidine, 2-thio-N4-methyl-5-hydroxy-5-azacytidine, 2-thio-N4-methyl-5-methylpseudoisocytidine, 2-thio-N4-methyl-5-aminopseudoisocytidine, 2-thio-N4-methyl-5-hydroxypseudoisocytidine, 2-thio-N4-amino-5-azacytidine, 2-thio-N4-aminopseudoisocytidine, 2-thio-N4-amino-5-methylcytidine, 2-thio-N4-amino-5-aminocytidine, 2-thio-N4-amino-5-hydroxycytidine, 2-thio-N4-amino-5-methyl-5-azacytidine, 2-thio-N4-amino-5-amino-5-azacytidine, 2-thio-N4-amino-5-hydroxy-5-azacytidine, 2-thio-N4-amino-5-methylpseudoisocytidine, 2-thio-N4-amino-5-aminopseudoisocytidine, 2-thio-N4-amino-5-hydroxypseudoisocytidine, 2-thio-N4-hydroxy-5-azacytidine, 2-thio-N4-hydroxypseudoisocytidine, 2-thio-N4-hydroxy-5-methylcytidine, N4-hydroxy-5-aminocytidine, 2-thio-N4-hydroxy-5-hydroxycytidine, 2-thio-N4-hydroxy-5-methyl-5-azacytidine, 2-thio-N4-hydroxy-5-amino-5-azacytidine, 2-thio-N4-hydroxy-5-hydroxy-5-azacytidine, 2-thio-N4-hydroxy-5-methylpseudoisocytidine, 2-thio-N4-hydroxy-5-aminopseudoisocytidine, 2-thio-N4-hydroxy-5-hydroxypseudoisocytidine, N6-methyladenosine, N6-aminoadenosine, N6-hydroxyadenosine, 7-deazaadenosine, 8-azaadenosine, N6-methyl-7-deazaadenosine, N6-methyl-8-azaadenosine, 7-deaza-8-azaadenosine, N6-methyl-7-deaza-8-azaadenosine, N6-amino-7-deazaadenosine, N6-amino-8-azaadenosine, N6-amino-7-deaza-8-azaadenosine, N6-hydroxyadenosine, N6-hydroxy-7-deazaadenosine, N6-hydroxy-8-azaadenosine, N6-hydroxy-7-deaza-8-azaadenosine, 6-thioguanosine, 7-deazaguanosine, 8-azaguanosine, 6-thio-7-deazaguanosine, 6-thio-8-azaguanosine, 7-deaza-8-azaguanosine, 6-thio-7-deaza-8-azaguanosine, and 5-methoxyuridine.
  • In some embodiments, the invention relates to one or more non-canonical nucleotides selected from 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, 5-hydroxyuridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine. In some embodiments, at least 50%, or at least 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of the non-canonical nucleotides are one or more of 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine.
  • In some embodiments, at least about 50%, or at least about 55%%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of cytidine residues are non-canonical nucleotides selected from 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine.
  • In some embodiments, at least about 20%, or about 30%, or about 40%, or about 50%, or at least about 55%, or at least 60%, or at least 65%, or at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or 100% of uridine residues are non-canonical nucleotides selected from 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine.
  • In some embodiments, at least about 10% (e.g. 10%, or about 20%, or about 30%, or about 40%, or about 50%) of guanosine residues are non-canonical nucleotides, and the non-canonical nucleotide is optionally 7-deazaguanosine. In some embodiments, the RNA contains no more than about 50% 7-deazaguanosine in place of guanosine residues.
  • In some embodiments, the RNA does not contain non-canonical nucleotides in place of adenosine residues.
  • Note that alternative naming schemes exist for certain non-canonical nucleotides. For example, in certain situations, 5-methylpseudouridine can be referred to as “3-methylpseudouridine” or “N3-methylpseudouridine” or “1-methylpseudouridine” or “N1-methylpseudouridine”.
  • Nucleotides that contain the prefix “amino” can refer to any nucleotide that contains a nitrogen atom bound to the atom at the stated position of the nucleotide, for example, 5-aminocytidine can refer to 5-aminocytidine, 5-methylaminocytidine, and 5-nitrocytidine. Similarly, nucleotides that contain the prefix “methyl” can refer to any nucleotide that contains a carbon atom bound to the atom at the stated position of the nucleotide, for example, 5-methylcytidine can refer to 5-methylcytidine, 5-ethylcytidine, and 5-hydroxymethylcytidine, nucleotides that contain the prefix “thio” can refer to any nucleotide that contains a sulfur atom bound to the atom at the given position of the nucleotide, and nucleotides that contain the prefix “hydroxy” can refer to any nucleotide that contains an oxygen atom bound to the atom at the given position of the nucleotide, for example, 5-hydroxyuridine can refer to 5-hydroxyuridine and uridine with a methyl group bound to an oxygen atom, wherein the oxygen atom is bound to the atom at the 5C position of the uridine.
  • Certain embodiments are therefore directed to RNA comprising one or more non-canonical nucleotides, wherein the RNA molecule contains one or more nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine. Other embodiments are directed to a therapeutic, wherein the therapeutic contains one or more RNA molecules comprising one or more non-canonical nucleotides, and wherein the one or more RNA molecules comprising one or more non-canonical nucleotides contains one or more nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine. In one embodiment, the therapeutic comprises a transfection reagent. In another embodiment, the transfection reagent comprises a cationic lipid, liposome or micelle. In still another embodiment, the liposome or micelle comprises folate and the therapeutic composition has anti-cancer activity. In another embodiment, the one or more nucleotides includes at least one of pseudouridine, 2-thiouridine, 4-thiouridine, 5-azauridine, 5-hydroxyuridine, 5-methyluridine, 5-aminouridine, 2-thiopseudouridine, 4-thiopseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-aminopseudouridine, pseudoisocytidine, N4-methylcytidine, 2-thiocytidine, 5-azacytidine, 5-hydroxycytidine, 5-aminocytidine, 5-methylcytidine, N4-methylpseudoisocytidine, 2-thiopseudoisocytidine, 5-hydroxypseudoisocytidine, 5-aminopseudoisocytidine, 5-methylpseudoisocytidine, 7-deazaadenosine, 7-deazaguanosine, 6-thioguanosine, and 6-thio-7-deazaguanosine. In another embodiment, the one or more nucleotides includes at least one of pseudouridine, 2-thiouridine, 4-thiouridine, 5-azauridine, 5-hydroxyuridine, 5-methyluridine, 5-aminouridine, 2-thiopseudouridine, 4-thiopseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, and 5-aminopseudouridine and at least one of pseudoisocytidine, N4-methylcytidine, 2-thiocytidine, 5-azacytidine, 5-hydroxycytidine, 5-aminocytidine, 5-methylcytidine, N4-methylpseudoisocytidine, 2-thiopseudoisocytidine, 5-hydroxypseudoisocytidine, 5-aminopseudoisocytidine, and 5-methylpseudoisocytidine. In still another embodiment, the one or more nucleotides include at least one of pseudouridine, 2-thiouridine, 4-thiouridine, 5-azauridine, 5-hydroxyuridine, 5-methyluridine, 5-aminouridine, 2-thiopseudouridine, 4-thiopseudouridine, 5-hydroxypseudouridine, and 5-methylpseudouridine, 5-aminopseudouridine and at least one of pseudoisocytidine, N4-methylcytidine, 2-thiocytidine, 5-azacytidine, 5-hydroxycytidine, 5-aminocytidine, 5-methylcytidine, N4-methylpseudoisocytidine, 2-thiopseudoisocytidine, 5-hydroxypseudoisocytidine, 5-aminopseudoisocytidine, and 5-methylpseudoisocytidine and at least one of 7-deazaguanosine, 6-thioguanosine, 6-thio-7-deazaguanosine, and 5-methoxyuridine. In yet another embodiment, the one or more nucleotides includes: 5-methylcytidine and 7-deazaguanosine. In another embodiment, the one or more nucleotides also includes pseudouridine or 4-thiouridine or 5-methyluridine or 5-aminouridine or 4-thiopseudouridine or 5-methylpseudouridine or 5-aminopseudouridine. In a still another embodiment, the one or more nucleotides also includes 7-deazaadenosine. In another embodiment, the one or more nucleotides includes: pseudoisocytidine and 7-deazaguanosine and 4-thiouridine. In yet another embodiment, the one or more nucleotides includes: pseudoisocytidine or 7-deazaguanosine and pseudouridine. In still another embodiment, the one or more nucleotides includes: 5-methyluridine and 5-methylcytidine and 7-deazaguanosine. In a further embodiment, the one or more nucleotides includes: pseudouridine or 5-methylpseudouridine and 5-methylcytidine and 7-deazaguanosine. In another embodiment, the one or more nucleotides includes: pseudoisocytidine and 7-deazaguanosine and pseudouridine. In one embodiment, the RNA comprising one or more non-canonical nucleotides is present in vivo.
  • Certain non-canonical nucleotides can be incorporated more efficiently than other non-canonical nucleotides into RNA molecules by RNA polymerases that are commonly used for in vitro transcription, due in part to the tendency of these certain non-canonical nucleotides to participate in standard base-pairing interactions and base-stacking interactions, and to interact with the RNA polymerase in a manner similar to that in which the corresponding canonical nucleotide interacts with the RNA polymerase. As a result, certain nucleotide mixtures containing one or more non-canonical nucleotides can be beneficial in part because in vitro-transcription reactions containing these nucleotide mixtures can yield a large quantity of RNA. Certain embodiments are therefore directed to a nucleotide mixture containing one or more nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine. Nucleotide mixtures include, but are not limited to (numbers preceding each nucleotide indicate an exemplary fraction of the non-canonical nucleotide triphosphate in an in vitro-transcription reaction, for example, 0.2 pseudoisocytidine refers to a reaction containing adenosine-5′-triphosphate, guanosine-5′-triphosphate, uridine-5′-triphosphate, cytidine-5′-triphosphate, and pseudoisocytidine-5′-triphosphate, wherein pseudoisocytidine-5′-triphosphate is present in the reaction at an amount approximately equal to 0.2 times the total amount of pseudoisocytidine-5′-triphosphate+cytidine-5′-triphosphate that is present in the reaction, with amounts measured either on a molar or mass basis, and wherein more than one number preceding a nucleoside indicates a range of exemplary fractions): 1.0 pseudouridine, 0.1-0.8 2-thiouridine, 0.1-0.8 5-methyluridine, 0.2-1.0 5-hydroxyuridine, 0.2-1.0 5-methoxyuridine, 0.1-1.0 5-aminouridine, 0.1-1.0 4-thiouridine, 0.1-1.0 2-thiopseudouridine, 0.1-1.0 4-thiopseudouridine, 0.1-1.0 5-hydroxypseudouridine, 0.2-1 5-methylpseudouridine, 0.2-1.0 5-methoxypseudouridine, 0.1-1.0 5-aminopseudouridine, 0.2-1.0 2-thiocytidine, 0.1-0.8 pseudoisocytidine, 0.2-1.0 5-methylcytidine, 0.2-1.0 5-hydroxycytidine, 0.2-1.0 5-hydroxymethylcytidine, 0.2-1.0 5-methoxycytidine, 0.1-1.0 5-aminocytidine, 0.2-1.0 N4-methylcytidine, 0.2-1.0 5-methylpseudoisocytidine, 0.2-1.0 5-hydroxypseudoisocytidine, 0.2-1.0 5-aminopseudoisocytidine, 0.2-1.0 N4-methylpseudoisocytidine, 0.2-1.0 2-thiopseudoisocytidine, 0.2-1.0 7-deazaguanosine, 0.2-1.0 6-thioguanosine, 0.2-1.0 6-thio-7-deazaguanosine, 0.2-1.0 8-azaguanosine, 0.2-1.0 7-deaza-8-azaguanosine, 0.2-1.0 6-thio-8-azaguanosine, 0.1-0.5 7-deazaadenosine, and 0.1-0.5 N6-methyladenosine.
  • In various embodiments, the RNA comprising one or more non-canonical nucleotides composition or synthetic polynucleotide composition (e.g., which may be prepared by in vitro transcription) contains substantially or entirely the canonical nucleotide at positions having adenine or “A” in the genetic code. The term “substantially” in this context refers to at least 90%. In these embodiments, the RNA composition or synthetic polynucleotide composition may further contain (e.g., consist of) 7-deazaguanosine at positions with “G” in the genetic code as well as the corresponding canonical nucleotide “G”, and the canonical and non-canonical nucleotide at positions with G may be in the range of 5:1 to 1:5, or in some embodiments in the range of 2:1 to 1:2. In these embodiments, the RNA composition or synthetic polynucleotide composition may further contain (e.g., consist of) one or more (e.g., two, three or four) of 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine at positions with “C” in the genetic code as well as the canonical nucleotide “C”, and the canonical and non-canonical nucleotide at positions with C may be in the range of 5:1 to 1:5, or in some embodiments in the range of 2:1 to 1:2. In some embodiments, the level of non-canonical nucleotide at positions of “C” are as described in the preceding paragraph. In these embodiments, the RNA composition or synthetic polynucleotide composition may further contain (e.g., consist of) one or more (e.g., two, three, or four) of 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, pseudouridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine at positions with “U” in the genetic code as well as the canonical nucleotide “U”, and the canonical and non-canonical nucleotide at positions with “U” may be in the range of 5:1 to 1:5, or in some embodiments in the range of 2:1 to 1:2. In some embodiments, the level of non-canonical nucleotide at positions of “U” are as described in the preceding paragraph.
  • It has now been discovered that combining certain non-canonical nucleotides can be beneficial in part because the contribution of non-canonical nucleotides to lowering the toxicity of RNA molecules can be additive. Certain embodiments are therefore directed to a nucleotide mixture, wherein the nucleotide mixture contains more than one of the non-canonical nucleotides listed above, for example, the nucleotide mixture contains both pseudoisocytidine and 7-deazaguanosine or the nucleotide mixture contains both N4-methylcytidine and 7-deazaguanosine, etc. In one embodiment, the nucleotide mixture contains more than one of the non-canonical nucleotides listed above, and each of the non-canonical nucleotides is present in the mixture at the fraction listed above, for example, the nucleotide mixture contains 0.1-0.8 pseudoisocytidine and 0.2-1.0 7-deazaguanosine or the nucleotide mixture contains 0.2-1.0 N4-methylcytidine and 0.2-1.0 7-deazaguanosine, etc.
  • In certain situations, for example, when it may not be necessary or desirable to maximize the yield of an in vitro-transcription reaction, nucleotide fractions other than those given above may be used. The exemplary fractions and ranges of fractions listed above relate to nucleotide-triphosphate solutions of typical purity (greater than 90% purity). Larger fractions of these and other nucleotides can be used by using nucleotide-triphosphate solutions of greater purity, for example, greater than about 95% purity or greater than about 98% purity or greater than about 99% purity or greater than about 99.5% purity, which can be achieved, for example, by purifying the nucleotide triphosphate solution using existing chemical-purification technologies such as high-pressure liquid chromatography (HPLC) or by other means. In one embodiment, nucleotides with multiple isomers are purified to enrich the desired isomer.
  • Other embodiments are directed to a method for inducing a cell in vivo to express a protein of interest by contacting the cell with a RNA molecule that contains one or more non-canonical nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine. Still other embodiments are directed to a method for transfecting, reprogramming, and/or gene-editing a cell in vivo by contacting the cell with a RNA molecule that contains one or more non-canonical nucleotides that includes one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine. In one embodiment, the RNA molecule is produced by in vitro transcription. In one embodiment, the RNA molecule encodes one or more reprogramming factors. In another embodiment, the one or more reprogramming factors includes Oct4 protein. In another embodiment, the cell is also contacted with a RNA molecule that encodes Sox2 protein. In yet another embodiment, the cell is also contacted with a RNA molecule that encodes Klf4 protein. In yet another embodiment, the cell is also contacted with a RNA molecule that encodes c-Myc protein. In yet another embodiment, the cell is also contacted with a RNA molecule that encodes Lin28 protein.
  • Enzymes such as T7 RNA polymerase may preferentially incorporate canonical nucleotides in an in vitro-transcription reaction containing both canonical and non-canonical nucleotides. As a result, an in vitro-transcription reaction containing a certain fraction of a non-canonical nucleotide may yield RNA containing a different, often lower, fraction of the non-canonical nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction. In certain embodiments, references to nucleotide incorporation fractions (for example, “a synthetic RNA molecule containing 50% pseudoisocytidine” or “0.1-0.8 pseudoisocytidine”) therefore can refer both to RNA molecules containing the stated fraction of the nucleotide, and to RNA molecules synthesized in a reaction containing the stated fraction of the nucleotide (or nucleotide derivative, for example, nucleotide-triphosphate), even though such a reaction may yield RNA containing a different fraction of the nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction.
  • Different nucleotide sequences can encode the same protein by utilizing alternative codons. In certain embodiments, references to nucleotide incorporation fractions therefore can refer both to RNA molecules containing the stated fraction of the nucleotide, and to RNA molecules encoding the same protein as a different RNA molecule, wherein the different RNA molecule contains the stated fraction of the nucleotide.
  • Certain embodiments are directed to a kit containing one or more materials needed to practice the present invention. In one embodiment, the kit contains one or more synthetic RNA molecules. In one embodiment, the kit contains one or more synthetic RNA molecules that encode one or more reprogramming factors and/or gene-editing proteins. In another embodiment, the one or more synthetic RNA molecules contain one or more non-canonical nucleotides that include one or more substitutions at the 2C and/or 4C and/or 5C positions in the case of a pyrimidine or the 6C and/or 7N and/or 8C positions in the case of a purine. In another embodiment, the kit contains one or more of: a transfection medium, a transfection reagent, a complexation medium, and a matrix solution. In one embodiment, the matrix solution contains fibronectin and/or vitronectin or recombinant fibronectin and/or recombinant vitronectin. In one embodiment, one or more of the components of the kit are present as a plurality of aliquots. In one embodiment, the kit contains aliquots of nucleic acid transfection-reagent complexes. In another embodiment, the kit contains aliquots of nucleic acid transfection-reagent complexes that are provided in a solid form, for example, as frozen or freeze-dried pellets. In yet another embodiment, the kit contains aliquots of medium, wherein each aliquot contains transfection reagent-nucleic acid complexes that are stabilized either by chemical treatment or by freezing.
  • Transfection, in general, and reprogramming, in particular, can be difficult and time-consuming techniques that can be repetitive and prone to error. However, these techniques are often performed manually due to the lack of automated transfection equipment. Certain embodiments are therefore directed to a system that can transfect, reprogram, and/or gene-edit cells in vivo in an automated or semi-automated manner.
  • It has now been discovered that the non-canonical nucleotide members of the 5-methylcytidine de-methylation pathway, when incorporated into synthetic RNA, can increase the efficiency with which the synthetic RNA can be translated into protein in vivo, and can decrease the toxicity of the synthetic RNA in vivo. These non-canonical nucleotides include, for example: 5-methylcytidine, 5-hydroxymethylcytidine, 5-formylcytidine, and 5-carboxycytidine (a.k.a. “cytidine-5-carboxylic acid”). Certain embodiments are therefore directed to a nucleic acid.
  • In some embodiments, the nucleic acid is present in vivo. In one embodiment, the nucleic acid is a synthetic RNA molecule. In another embodiment, the nucleic acid comprises one or more non-canonical nucleotides. In one embodiment, the nucleic acid comprises one or more non-canonical nucleotide members of the 5-methylcytidine de-methylation pathway. In another embodiment, the nucleic acid comprises at least one of: 5-methylcytidine, 5-hydroxymethylcytidine, 5-formylcytidine, and 5-carboxycytidine or a derivative thereof. In a further embodiment, the nucleic acid comprises at least one of: pseudouridine, 5-methylpseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, N4-methylcytidine, N4-acetylcytidine, and 7-deazaguanosine or a derivative thereof.
  • 5-methylcytidine De-Methylation Pathway
  • Figure US20200261599A1-20200820-C00002
  • Certain embodiments are directed to a protein. Other embodiments are directed to a nucleic acid that encodes a protein. In one embodiment, the protein is a protein of interest. In another embodiment, the protein is selected from: a reprogramming protein and a gene-editing protein. In one embodiment, the nucleic acid is a plasmid. In another embodiment, the nucleic acid is present in a virus or viral vector. In a further embodiment, the virus or viral vector is replication incompetent. In a still further embodiment, the virus or viral vector is replication competent. In one embodiment, the virus or viral vector includes at least one of: an adenovirus, a retrovirus, a lentivirus, a herpes virus, an adeno-associated virus or a natural or engineered variant thereof, and an engineered virus.
  • It has also been discovered that certain combinations of non-canonical nucleotides can be particularly effective at increasing the efficiency with which synthetic RNA can be translated into protein in vivo, and decreasing the toxicity of synthetic RNA in vivo, for example, the combinations: 5-methyluridine and 5-methylcytidine, 5-hydroxyuridine and 5-methylcytidine, 5-hydroxyuridine and 5-hydroxymethylcytidine, 5-methyluridine and 7-deazaguanosine, 5-methylcytidine and 7-deazaguanosine, 5-methyluridine, 5-methylcytidine, and 7-deazaguanosine, and 5-methyluridine, 5-hydroxymethylcytidine, and 7-deazaguanosine. Certain embodiments are therefore directed to a nucleic acid comprising at least two of: 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, and 7-deazaguanosine or one or more derivatives thereof. Other embodiments are directed to a nucleic acid comprising at least three of: 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, and 7-deazaguanosine or one or more derivatives thereof. Other embodiments are directed to a nucleic acid comprising all of: 5-methyluridine, 5-methylcytidine, 5-hydroxymethylcytidine, and 7-deazaguanosine or one or more derivatives thereof. In one embodiment, the nucleic acid comprises one or more 5-methyluridine residues, one or more 5-methylcytidine residues, and one or more 7-deazaguanosine residues or one or more 5-methyluridine residues, one or more 5-hydroxymethylcytidine residues, and one or more 7-deazaguanosine residues.
  • It has been further discovered that synthetic RNA molecules containing certain fractions of certain non-canonical nucleotides and combinations thereof can exhibit particularly high translation efficiency and low toxicity in vivo. Certain embodiments are therefore directed to a nucleic acid comprising at least one of: one or more uridine residues, one or more cytidine residues, and one or more guanosine residues, and comprising one or more non-canonical nucleotides. In one embodiment, between about 20% and about 80% of the uridine residues are 5-methyluridine residues. In another embodiment, between about 30% and about 50% of the uridine residues are 5-methyluridine residues. In a further embodiment, about 40% of the uridine residues are 5-methyluridine residues. In one embodiment, between about 60% and about 80% of the cytidine residues are 5-methylcytidine residues. In another embodiment, between about 80% and about 100% of the cytidine residues are 5-methylcytidine residues. In a further embodiment, about 100% of the cytidine residues are 5-methylcytidine residues. In a still further embodiment, between about 20% and about 100% of the cytidine residues are 5-hydroxymethylcytidine residues. In one embodiment, between about 20% and about 80% of the guanosine residues are 7-deazaguanosine residues. In another embodiment, between about 40% and about 60% of the guanosine residues are 7-deazaguanosine residues. In a further embodiment, about 50% of the guanosine residues are 7-deazaguanosine residues. In one embodiment, between about 20% and about 80% or between about 30% and about 60% or about 40% of the cytidine residues are N4-methylcytidine and/or N4-acetylcytidine residues. In another embodiment, each cytidine residue is a 5-methylcytidine residue. In a further embodiment, about 100% of the cytidine residues are 5-methylcytidine residues and/or 5-hydroxymethylcytidine residues and/or N4-methylcytidine residues and/or N4-acetylcytidine residues and/or one or more derivatives thereof. In a still further embodiment, about 40% of the uridine residues are 5-methyluridine residues, between about 20% and about 100% of the cytidine residues are N4-methylcytidine and/or N4-acetylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues. In one embodiment, about 40% of the uridine residues are 5-methyluridine residues and about 100% of the cytidine residues are 5-methylcytidine residues. In another embodiment, about 40% of the uridine residues are 5-methyluridine residues and about 50% of the guanosine residues are 7-deazaguanosine residues. In a further embodiment, about 100% of the cytidine residues are 5-methylcytidine residues and about 50% of the guanosine residues are 7-deazaguanosine residues. In a further embodiment, about 100% of the uridine residues are 5-hydroxyuridine residues. In one embodiment, about 40% of the uridine residues are 5-methyluridine residues, about 100% of the cytidine residues are 5-methylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues. In another embodiment, about 40% of the uridine residues are 5-methyluridine residues, between about 20% and about 100% of the cytidine residues are 5-hydroxymethylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues. In some embodiments, less than 100% of the cytidine residues are 5-methylcytidine residues. In other embodiments, less than 100% of the cytidine residues are 5-hydroxymethylcytidine residues. In one embodiment, each uridine residue in the synthetic RNA molecule is a pseudouridine residue or a 5-methylpseudouridine residue. In another embodiment, about 100% of the uridine residues are pseudouridine residues and/or 5-methylpseudouridine residues. In a further embodiment, about 100% of the uridine residues are pseudouridine residues and/or 5-methylpseudouridine residues, about 100% of the cytidine residues are 5-methylcytidine residues, and about 50% of the guanosine residues are 7-deazaguanosine residues.
  • Other non-canonical nucleotides that can be used in place of or in combination with 5-methyluridine include, but are not limited to: pseudouridine, 5-hydroxyuridine, 5-hydroxypseudouridine, 5-methoxyuridine, 5-methoxypseudouridine, 5-carboxyuridine, 5-carboxypseudouridine, 5-formyluridine, 5-formylpseudouridine, 5-hydroxymethyluridine, 5-hydroxymethylpseudouridine, and 5-methylpseudouridine (a.k.a. “1-methylpseudouridine”, a.k.a. “N1-methylpseudouridine”) or one or more derivatives thereof. Other non-canonical nucleotides that can be used in place of or in combination with 5-methylcytidine and/or 5-hydroxymethylcytidine include, but are not limited to: pseudoisocytidine, 5-methylpseudoisocytidine, 5-hydroxymethylcytidine, 5-formylcytidine, 5-carboxycytidine, 5-methoxycytidine, N4-methylcytidine, N4-acetylcytidine or one or more derivatives thereof. In certain embodiments, for example, when performing only a single transfection, injection or delivery or when the cells, tissue, organ or patient being transfected, injected or delivered to are not particularly sensitive to transfection-associated toxicity or innate-immune signaling, the fractions of non-canonical nucleotides can be reduced. Reducing the fraction of non-canonical nucleotides can be beneficial, in part, because reducing the fraction of non-canonical nucleotides can reduce the cost of the nucleic acid. In certain situations, for example, when minimal immunogenicity of the nucleic acid is desired, the fractions of non-canonical nucleotides can be increased.
  • Enzymes such as T7 RNA polymerase may preferentially incorporate canonical nucleotides in an in vitro-transcription reaction containing both canonical and non-canonical nucleotides. As a result, an in vitro-transcription reaction containing a certain fraction of a non-canonical nucleotide may yield RNA containing a different, often lower, fraction of the non-canonical nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction. In certain embodiments, references to nucleotide incorporation fractions (for example, “50% 5-methyluridine”) therefore can refer both to nucleic acids containing the stated fraction of the nucleotide, and to nucleic acids synthesized in a reaction containing the stated fraction of the nucleotide (or nucleotide derivative, for example, nucleotide-triphosphate), even though such a reaction may yield a nucleic acid containing a different fraction of the nucleotide than the fraction at which the non-canonical nucleotide was present in the reaction. In addition, different nucleotide sequences can encode the same protein by utilizing alternative codons. In certain embodiments, references to nucleotide incorporation fractions therefore can refer both to nucleic acids containing the stated fraction of the nucleotide, and to nucleic acids encoding the same protein as a different nucleic acid, wherein the different nucleic acid contains the stated fraction of the nucleotide.
  • Certain embodiments are directed to a nucleic acid comprising a 5′-cap structure selected from Cap 0, Cap 1, Cap 2, and Cap 3 or a derivative thereof. In one embodiment, the nucleic acid comprises one or more UTRs. In another embodiment, the one or more UTRs increase the stability of the nucleic acid. In a further embodiment, the one or more UTRs comprise an alpha-globin or beta-globin 5′-UTR. In a still further embodiment, the one or more UTRs comprise an alpha-globin or beta-globin 3′-UTR. In a still further embodiment, the synthetic RNA molecule comprises an alpha-globin or beta-globin 5′-UTR and an alpha-globin or beta-globin 3′-UTR. In one embodiment, the 5′-UTR comprises a Kozak sequence that is substantially similar to the Kozak consensus sequence. In another embodiment, the nucleic acid comprises a 3′-poly(A) tail. In a further embodiment, the 3′-poly(A) tail is between about 20 nt and about 250 nt or between about 120 nt and about 150 nt long. In a further embodiment, the 3′-poly(A) tail is about 20 nt, or about 30 nt, or about 40 nt, or about 50 nt, or about 60 nt, or about 70 nt, or about 80 nt, or about 90 nt, or about 100 nt, or about 110 nt, or about 120 nt, or about 130 nt, or about 140 nt, or about 150 nt, or about 160 nt, or about 170 nt, or about 180 nt, or about 190 nt, or about 200 nt, or about 210 nt, or about 220 nt, or about 230 nt, or about 240 nt, or about 250 nt long.
  • Certain embodiments are directed to methods of making nucleic acid drugs, including RNA comprising one or more non-canonical nucleotides. Such methods yield substantially stable RNA.
  • In various embodiments, the present methods and compositions find use in methods of treating, preventing or ameliorating a disease, disorder and/or condition. For instance, in some embodiments, the described methods of in vivo delivery, including various effective doses, administration strategies and formulations are used in a method of treatment.
  • In various embodiments, the present methods and compositions find use in methods of altering, modifying and/or changing a tissue (e.g. cosmetically).
  • In various embodiments, the present methods and compositions include using a nucleic acid drug, including a synthetic RNA, in the diagnosing, treating, preventing or ameliorating of a disease, disorder and/or condition described herein. In various embodiments, the present methods and compositions include using a nucleic acid drug, including a synthetic RNA, in the altering, modifying and/or changing of a tissue (e.g. cosmetically).
  • Generally speaking, in various embodiments, a synthetic RNA as described herein is administered to a human at specific doses described herein and the synthetic RNA comprises a sequence, sometimes referred to as a target sequence that encodes a protein of interest, which may be a therapeutic protein.
  • Synthetic RNA comprising only canonical nucleotides can bind to pattern recognition receptors, can be recognized as a pathogen-associated molecular pattern, and can trigger a potent immune response in cells, which can result in translation block, the secretion of inflammatory cytokines, and cell death. It has now been discovered that synthetic RNA comprising certain non-canonical nucleotides can evade detection by the innate immune system, and can be translated at high efficiency into protein, including in humans. It has been further discovered that synthetic RNA comprising at least one of the non-canonical nucleotides described herein, including, for example, a member of the group: 5-methylcytidine, 5-hydroxycytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-methoxyuridine, 5-formyluridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-methoxypseudouridine, and 5-formylpseudouridine can evade detection by the innate immune system, and can be translated at high efficiency into protein, including in humans. Certain embodiments are therefore directed to a method for inducing a cell to express a protein of interest comprising contacting a cell with synthetic RNA. Other embodiments are directed to a method for transfecting a cell with synthetic RNA comprising contacting a cell with a solution comprising one or more synthetic RNA molecules. Still other embodiments are directed to a method for treating a patient comprising administering to the patient synthetic RNA. In one embodiment, the synthetic RNA comprises at least one of the non-canonical nucleotides described herein, including, for example, a member of the group: 5-methylcytidine, 5-hydroxycytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-methoxyuridine, 5-formyluridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-methoxypseudouridine, and 5-formylpseudouridine. In another embodiment, the synthetic RNA encodes a protein of interest. Exemplary RNAs may contain combinations and levels of non-canonical and non-canonical nucleotides as described elsewhere herein, including with respect to the expression of any protein of interest described herein. In yet another embodiment, the method results in the expression of the protein of interest. In a further embodiment, the method results in the expression of the protein of interest in the patient's skin.
  • Other embodiments are directed to a method for delivering a nucleic acid to a cell in vivo. Still other embodiments are directed to a method for inducing a cell in vivo to express a protein of interest. Still other embodiments are directed to a method for treating a patient. In one embodiment, the method comprises disrupting the stratum corneum. In another embodiment, the method comprises contacting a cell with a nucleic acid. In yet another embodiment, the method results in the cell internalizing the nucleic acid. In a further embodiment, the method results in the cell expressing the protein of interest. In a still further embodiment, the method results in the expression of the protein of interest in the patient. In a still further embodiment, the method results in the amelioration of one or more of the patient's symptoms. In a still further embodiment, the patient is in need of the protein of interest. In a still further embodiment, the patient is deficient in the protein of interest.
  • Still other embodiments are directed to a method for treating a patient comprising delivering to a patient a composition. In one embodiment, the composition comprises albumin that is treated with an ion-exchange resin or charcoal. In another embodiment, the composition comprises one or more nucleic acid molecules. In yet another embodiment, at least one of the one or more nucleic acid molecules encodes a protein of interest. In one embodiment, the method results in the expression of the protein in the patient's skin. In another embodiment, the method results in the expression of a therapeutically or cosmetically effective amount of the protein of interest in the patient. In yet another embodiment, the method comprises administering a steroid. In a further embodiment, the steroid is a member of the group: hydrocortisone and dexamethasone.
  • Some embodiments are directed to a therapeutic composition and/or methods of treatment comprising a nucleic acid molecule encoding one or more proteins, wherein at least one of the one or more proteins is an extracellular matrix protein. Still other embodiments are directed to a cosmetic composition comprising a nucleic acid molecule encoding one or more proteins, wherein at least one of the one or more proteins is an extracellular matrix protein.
  • Pigmentation disorders can cause severe symptoms in patients. It has now been discovered that pigmentation disorders can be treated by delivering to a patient a nucleic acid encoding tyrosinase. Certain embodiments are therefore directed to a method for treating a pigmentation disorder. Other embodiments are directed to a method for altering the pigmentation of a patient. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding tyrosinase. Other embodiments are directed to a cosmetic composition comprising a nucleic acid encoding tyrosinase. Still other embodiments are directed to a therapeutic composition comprising a nucleic acid encoding tyrosinase. Still other embodiments are directed to a method for increasing the ultraviolet absorption of a patient's skin. In one embodiment the method comprises delivering to a patient a nucleic acid encoding tyrosinase. In another embodiment, the method results in an increase in the ultraviolet absorption of the patient's skin. Still other embodiments are directed to a method for reducing photodamage to a person's skin upon exposure to ultraviolet light. In one embodiment, the method results in the reduction of photodamage to the person's skin upon exposure to ultraviolet light. Still other embodiments are directed to a method for treating xeroderma pigmentosum. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding tyrosinase. Still other embodiments are directed to a method for treating epidermolysis bullosa. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding one or more of keratin 5, keratin 14, plectin, an integrin family member, laminin, a laminin subunit, collagen XVII, collagen VII or a biologically active fragment, variant, analogue or family-member thereof. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding collagen type VII. In another embodiment, the method comprises delivering to a patient a nucleic acid encoding melanocortin 1 receptor. Still other embodiments are directed to a method for treating xerosis. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding a hyaluronan synthase. In another embodiment, the patient is diagnosed with atopic dermatitis. In yet another embodiment, the patient is diagnosed with ichthyosis. Certain embodiments are directed to a method for treating a cosmetic condition. Other embodiments are directed to a method for inducing tissue healing. In one embodiment, the method comprises delivering to a patient a nucleic acid encoding a hyaluronan synthase. In another embodiment, the cosmetic condition is a member of the group: wrinkles, sagging skin, thin skin, discoloration, and dry skin. In yet another embodiment, the patient has had cataract surgery. In some embodiments, the nucleic acid is synthetic RNA. In other embodiments, the method results in the amelioration of one or more of the patient's symptoms. Other embodiments are directed to a method for treating an indication by delivering to a cell or a patient a nucleic acid encoding a protein or a peptide. Still other embodiments are directed to a composition comprising a nucleic acid encoding a protein or a peptide. Indications that can be treated using the methods and compositions of the present invention and proteins and peptides that can be encoded by compositions of the present invention are set forth in Table 2A and/or Table 2B, and are given by way of example, and not by way of limitation. In one embodiment, the indication is selected from Table 2A and/or Table 2B. In another embodiment the protein or peptide is selected from Table 2A and/or Table 2B. In yet another embodiment, the indication and the protein or peptide are selected from the same row of Table 2A and/or Table 2B. In a further embodiment, the protein of interest is a member of the group: UCP1, UCP2, and UCP3. Other embodiments are directed to methods for inducing a cell to express a plurality of proteins of interest. In one embodiment, the proteins of interest include at least two members of the group: a lipase, UCP1, UCP2, and UCP3. In another embodiment, the proteins of interest include a lipase and a member of the group: UCP1, UCP2, and UCP3. In another embodiment, the protein is a gene-editing protein. In yet another embodiment, the gene-editing protein targets a gene that is at least partly responsible for a disease phenotype. In yet another embodiment, the gene-editing protein targets a gene that encodes a protein selected from Table 2A and/or Table 2B. In still another embodiment, the gene-editing protein corrects or eliminates, either alone or in combination with one or more other molecules or gene-editing proteins, a mutation that is at least partly responsible for a disease phenotype.
  • In various embodiments, the present invention contemplates the targeting of the precursor forms and/or mature forms and/or isoforms and/or mutants of any of the proteins disclosed in Table 2A and/or Table 2B and such proteins. In some embodiments, any of the precursor forms and/or mature forms and/or isoforms and/or mutants have enhanced secretion relative to the corresponding wild type proteins. In some embodiments, any of the precursor forms and/or mature forms and/or isoforms and/or mutants have altered half-lives (e.g. serum, plasma, intracellular)—for instance, longer or shorter half-lives. In some embodiments, this is relative to wild type.
  • TABLE 2A
    Illustrative Indications
    Illustrative Indication Illustrative Protein/Peptide
    Acne Retinol Dehydrogenase 10
    Aging Elastin
    sp|P15502|ELN_HUMAN Elastin
    (isoform 3)
    (SEQ ID NO: 486)
    Aging Collagen Type I
    P02452|CO1A1_HUMAN Collagen alpha-1(I) chain
    (SEQ ID NO: 487)
    P08123|CO1A2_HUMAN Collagen alpha-2(I) chain
    (SEQ ID NO: 488)
    Aging Collagen Type III
    P02461|CO3A1_HUMAN Collagen alpha-1(III) chain
    (isoform 1)
    (SEQ ID NO: 489)
    Aging Collagen Type VII
    Q02388|CO7A1_HUMAN Collagen alpha-1(VII)
    chain
    (SEQ ID NO: 490)
    Aging Hyaluronan Synthase
    Aging Telomerase Reverse Transcriptase
    Albinism Tyrosinase
    P14679|TYRO_HUMAN Tyrosinase
    (isoform 1)
    (SEQ ID NO: 491)
    Alport Syndrome Collagen Type IV
    P02462|CO4A1_HUMAN Collagen alpha-1(IV) chain
    (isoform 1)
    (SEQ ID NO: 492)
    P08572|CO4A2_HUMAN Collagen alpha-2(IV) chain
    (SEQ ID NO: 493)
    Q01955|CO4A3_HUMAN Collagen alpha-3(IV) chain
    (isoform 1)
    (SEQ ID NO: 494)
    P53420|CO4A4_HUMAN Collagen alpha-4(IV) chain
    (SEQ ID NO: 495)
    P29400|CO4A5_HUMAN Collagen alpha-5(IV) chain
    (isoform 1)
    (SEQ ID NO: 496)
    Q14031|CO4A6_HUMAN Collagen alpha-6(IV)
    (isoform A)
    (SEQ ID NO: 497)
    Anemia Erythropoietin
    Atopic Dermatitis Filaggrin
    Cutis Laxa Elastin
    sp|P15502|ELN_HUMAN Elastin
    (isoform 3)
    (SEQ ID NO: 486)
    Dry Skin Filaggrin
    Dystrophic Epidermolysis Bullosa Collagen Type VII
    Q02388|CO7A1_HUMAN Collagen alpha-1(VII)
    chain
    (SEQ ID NO: 498)
    Ehlers-Danlos Syndrome Collagen Type V
    P20908|CO5A1_HUMAN Collagen alpha-1(V) chain
    (SEQ ID NO: 499)
    P05997|CO5A2_HUMAN Collagen alpha-2(V) chain
    (SEQ ID NO: 500)
    P25940|CO5A3_HUMAN Collagen alpha-3(V) chain
    (SEQ ID NO: 501)
    Ehlers-Danlos Syndrome Collagen Type I
    P02452|CO1A1_HUMAN Collagen alpha-1(I) chain
    (SEQ ID NO: 487)
    P08123|CO1A2_HUMAN Collagen alpha-2(I) chain
    (SEQ ID NO: 488)
    Epidermolysis bullosa, lethal acantholytic ADAM17
    P78536|ADA17_HUMAN Disintegrin and
    metalloproteinase domain-containing protein 17
    (isoform A)
    (SEQ ID NO: 502)
    Epidermolysis bullosa, type IV Collagen Type III
    P02461|CO3A1_HUMAN Collagen alpha-1(III) chain
    (isoform 1)
    (SEQ ID NO: 489)
    Erythropoietic Protoporphyria Ferrochelatase
    P22830|HEMH_HUMAN Ferrochelatase,
    mitochondrial
    (isoform 1)
    (SEQ ID NO: 503)
    Eczema Filaggrin
    Excess Fat Thermogenin
    P25874|UCP1_HUMAN Mitochondrial brown fat
    uncoupling protein 1
    (SEQ ID NO: 504)
    Excess Fat Lipase
    Lipoprotein lipase
    P06858|LIPL_HUMAN Lipoprotein lipase
    (SEQ ID NO: 516)
    Hepatic lipase
    P11150|LIPC_HUMAN Hepatic triacylglycerol lipase
    (SEQ ID NO: 517)
    Pancreatic lipase
    P16233|LIPP_HUMAN Pancreatic triacylglycerol
    lipase
    (SEQ ID NO: 518)
    Endothelial lipase
    (isoform 1)
    Q9Y5X9|LIPE_HUMAN Endothelial lipase
    (SEQ ID NO: 519)
    Lysosomal lipase
    P38571|LICH_HUMAN Lysosomal acid
    lipase/cholesteryl ester hydrolase
    (isoform 1)
    (SEQ ID NO: 520)
    Hormone sensitive lipase
    Q05469|LIPS_HUMAN Hormone-sensitive lipas
    (isoform 1)
    (SEQ ID NO: 521)
    Gastric lipase
    P07098|LIPG_HUMAN Gastric triacylglycerol lipase
    (isoform 1)
    (SEQ ID NO: 522)
    Pancreatic Lipase-Related Protein 1)
    P54315|LIPR1_HUMAN Inactive pancreatic lipase-
    related protein 1
    (isoform 1)
    (SEQ ID NO: 523)
    Pancreatic Lipase-Related Protein 2
    P54317|LIPR2_HUMAN Pancreatic lipase-related
    protein 2
    (SEQ ID NO: 524)
    Carboxyl Ester Lipase
    P19835|CEL_HUMAN Bile salt-activated lipase
    (isoform long)
    (SEQ ID NO: 525)
    Hypotrichosis ADAM17
    P78536|ADA17_HUMAN Disintegrin and
    metalloproteinase domain-containing protein 17
    (isoform A)
    (SEQ ID NO: 502)
    Ichthyosis Vulgaris Filaggrin
    Infections Genetic Antibiotics (e.g. Anti-Sigma Factors)
    Inflammatory and Bullous Skin Bowel Syndrome Desmoglein 2
    Q14126|DSG2_HUMAN Desmoglein-2
    (SEQ ID NO: 505)
    Keratosis Pilaris Retinol Dehydrogenase 10
    Oily Skin Retinol Dehydrogenase 10
    Osteoarthritis Hyaluronan Synthase
    Pemphigus Vulgaris Plakophilin-1
    Q13835|PKP1_HUMAN Plakophilin-1
    (isoform 2)
    (SEQ ID NO: 506)
    Pseudoxanthoma elasticum Elastin
    sp|P15502|ELN_HUMAN Elastin
    (isoform 3)
    (SEQ ID NO: 486)
    Psoriasis Retinol Dehydrogenase 10
    Scar Treatment Tyrosinase
    P14679|TYRO_HUMAN Tyrosinase
    (isoform 1)
    (SEQ ID NO: 491)
    Scarring Elastin
    sp|P15502|ELN_HUMAN Elastin
    (isoform 3)
    (SEQ ID NO: 486)
    Scarring Collagen Type I
    P02452|CO1A1_HUMAN Collagen alpha-1(I) chain
    (SEQ ID NO: 487)
    P08123|CO1A2_HUMAN Collagen alpha-2(I) chain
    (SEQ ID NO: 488)
    Scarring Collagen Type III
    P02461|CO3A1_HUMAN Collagen alpha-1(III) chain
    (isoform 1)
    (SEQ ID NO: 489)
    Skin Cancer Interferon
    Interferon, Alpha 1
    P01562|IFNA1_HUMAN Interferon alpha-1/13
    (SEQ ID NO: 530)
    Interferon, Alpha 2
    P01563|IFNA2_HUMAN Interferon alpha-2
    (SEQ ID NO: 531)
    Interferon, Alpha 4
    P05014|IFNA4_HUMAN Interferon alpha-4
    (SEQ ID NO: 532)
    Interferon, Alpha 5
    P01569|IFNA5_HUMAN Interferon alpha-5
    (SEQ ID NO: 533)
    Interferon, Alpha 6
    P05013|IFNA6_HUMAN Interferon alpha-6
    (SEQ ID NO: 534)
    Interferon, Alpha 7
    P01567|IFNA7_HUMAN Interferon alpha-7
    (SEQ ID NO: 535)
    Interferon, Alpha 8
    P32881|IFNA8_HUMAN Interferon alpha-8
    (SEQ ID NO: 536)
    Interferon, Alpha 10
    P01566|IFN10_HUMAN Interferon alpha-10
    (SEQ ID NO: 537)
    Interferon, Alpha 14
    P01570|IFN14_HUMAN Interferon alpha-14 OS
    (SEQ ID NO: 538)
    Interferon, Alpha 16
    P05015|IFN16_HUMAN Interferon alpha-16
    (SEQ ID NO: 539)
    Interferon, Alpha 17
    P01571|IFN17_HUMAN Interferon alpha-17
    (SEQ ID NO: 540)
    Interferon, Alpha 21
    P01568|IFN21_HUMAN Interferon alpha-21
    (SEQ ID NO: 541)
    Interferon, Gamma
    P01579|IFNG_HUMAN Interferon gamma
    (SEQ ID NO: 542)
    Interferon, Beta
    P01574|IFNB_HUMAN Interferon beta
    (SEQ ID NO: 543)
    Interferon, Kappa
    Q9P0W0|IFNK_HUMAN Interferon kappa
    (SEQ ID NO: 544)
    Interferon, Epsilon
    Q86WN2|IFNE_HUMAN Interferon epsilon
    (SEQ ID NO: 545)
    Striate Palmoplantar Keratoderma ADAM17
    P78536|ADA17_HUMAN Disintegrin and
    metalloproteinase domain-containing protein 17
    (isoform A)
    (SEQ ID NO: 502)
    Tanning Tyrosinase
    P14679|TYRO_HUMAN Tyrosinase
    (isoform 1)
    (SEQ ID NO: 491)
    Vitiligo Melanocyte-Stimulating Hormone
    Alpha-MSH
    P01189|138-150
    (SEQ ID NO: 526)
    Beta-MSH
    P01189|217-234
    (SEQ ID NO: 527)
    Gamma-MSH
    P01189|77-87
    (SEQ ID NO: 528)
    Proopiomelanocortin
    P01189|COLI_HUMAN Pro-opiomelanocortin
    (SEQ ID NO: 529)
    Vitiligo Tyrosinase
    P14679|TYRO_HUMAN Tyrosinase
    (isoform 1)
    (SEQ ID NO: 491)
    Warts Interferon
    Interferon, Alpha 1
    P01562|IFNA1_HUMAN Interferon alpha-1/13
    (SEQ ID NO: 530)
    Interferon, Alpha 2
    P01563|IFNA2_HUMAN Interferon alpha-2
    (SEQ ID NO: 531)
    Interferon, Alpha 4
    P05014|IFNA4_HUMAN Interferon alpha-4
    (SEQ ID NO: 532)
    Interferon, Alpha 5
    P01569|IFNA5_HUMAN Interferon alpha-5
    (SEQ ID NO: 533)
    Interferon, Alpha 6
    P05013|IFNA6_HUMAN Interferon alpha-6
    (SEQ ID NO: 534)
    Interferon, Alpha 7
    P01567|IFNA7_HUMAN Interferon alpha-7
    (SEQ ID NO: 535)
    Interferon, Alpha 8
    P32881|IFNA8_HUMAN Interferon alpha-8
    (SEQ ID NO: 536)
    Interferon, Alpha 10
    P01566|IFN10_HUMAN Interferon alpha-10
    (SEQ ID NO: 537)
    Interferon, Alpha 14
    P01570|IFN14_HUMAN Interferon alpha-14 OS
    (SEQ ID NO: 538)
    Interferon, Alpha 16
    P05015|IFN16_HUMAN Interferon alpha-16
    (SEQ ID NO: 539)
    Interferon, Alpha 17
    P01571|IFN17_HUMAN Interferon alpha-17
    (SEQ ID NO: 540)
    Interferon, Alpha 21
    P01568|IFN21_HUMAN Interferon alpha-21
    (SEQ ID NO: 541)
    Interferon, Gamma
    P01579|IFNG_HUMAN Interferon gamma
    (SEQ ID NO: 542)
    Interferon, Beta
    P01574|IFNB_HUMAN Interferon beta
    (SEQ ID NO: 543)
    Interferon, Kappa
    Q9P0W0|IFNK_HUMAN Interferon kappa
    (SEQ ID NO: 544)
    Interferon, Epsilon
    Q86WN2|IFNE_HUMAN Interferon epsilon
    (SEQ ID NO: 545)
    Wound Healing Elastin
    sp|P15502|ELN_HUMAN Elastin
    (isoform 3)
    (SEQ ID NO: 486)
    Wound Healing Collagen Type I
    P02452|CO1A1_HUMAN Collagen alpha-1(I) chain
    (SEQ ID NO: 487)
    P08123|CO1A2_HUMAN Collagen alpha-2(I) chain
    (SEQ ID NO: 488)
    Wound Healing Collagen Type III
    P02461|CO3A1_HUMAN Collagen alpha-1(III) chain
    (isoform 1)
    (SEQ ID NO: 489)
    Xeroderma Pigmentosum DNA Polymerase Eta
    Q9Y253|POLH_HUMAN DNA polymerase eta
    (isoform 1)
    (SEQ ID NO: 507)
  • Additional illustrative targets of the present invention include the cosmetic targets listed in Table 6 of International Patent Publication No. WO 2013/151671, the contents of which are hereby incorporated by reference in their entirety.
  • In various embodiments, the agents of the present invention are used in methods the effect the integumentary system of a human. The present compositions and methods may be used to alter a biological and/or physiological process to, for example, reduce skin sagging, increase skin thickness, increase skin volume, reduce the number of wrinkles, the length of wrinkles and/or the depth of wrinkles, increase skin tightness, firmness, tone and/or elasticity, increase skin hydration and ability to retain moisture, water flow and osmotic balance, increase the levels of skin lipids; increase the extracellular matrix and/or adhesion and communication polypeptides; increase skin energy production; utilization and conservation; improve oxygen utilization; improve skin cell life; improve skin cell immunity defense, heat shock stress response, antioxidant defense capacity to neutralize free radicals, and/or toxic defense; improve the protection and recovery from ultraviolet rays; improve skin cell communication and skin cell innervations; improve cell cohesion/adhesion; improve calcium mineral and other mineral metabolism; improve cell turnover; and improve cell circadian rhythms.
  • Further still, in some embodiments, the present compositions may be used in the treatment, control, or prevention of a disease, disorder and/or condition and/or may alter, modify or change the appearance of a member of the integumentary system of a subject suffering from a disease, disorder and/or condition such as, but not limited to, acne vulgaris, acne aestivalis, acne conglobata, acne cosmetic, acne fulminans, acne keloidalis nuchae, acne mechanica, acne medicamentosa, acne miliaris necrotica, acne necrotica, acne rosacea, actinic keratosis, acne vulgaris, acne aestivalis, acne conglobata, acne cosmetic, acne fulminans, acne keloidalis nuchae, acne mechanica, acne medicamentosa, acne miliaris necrotica, acne necrotica, acne rosacea, acute urticaria, allergic contact dermatitis, alopecia areata, angioedema, athlete's foot, atopic dermatitis, autoeczematization, baby acne, balding, bastomycosis, blackheads, birthmarks and other skin pigmentation problems, boils, bruises, bug bites and stings, burns, cellulitis, chiggers, chloracne, cholinergic or stress uricara, chronic urticara, cold type urticara, confluent and reticulated papillomatosis, corns, cysts, dandruff, dermatitis herpetiformis, dermatographism, dyshidrotic eczema, diaper rash, dry skin, dyshidrosis, ectodermal dysplasia such as, hyprohidrotic ectodermal dysplasia and X-linked hyprohidrotic ectodermal dysplasia, eczema, epidermaodysplasia verruciformis, erythema nodosum, excoriated acne, exercise-induced anaphylasis folliculitis, excess skin oil, folliculitis, freckles, frostbite, fungal nails, hair density, hair growth rate, halogen acne, hair loss, heat rash, hematoma, herpes simplex infections (e.g. non-genital), hidradenitis suppurativa, hives, hyperhidrosis, hyperpigmentation, hypohidrotic ectodermal dysplasia, hypopigmentation, impetigo, ingrown hair, heat type urticara, ingrown toenail, infantile acne or neonatal acne, itch, irritant contact dermatitis, jock itch, keloid, keratosis pilaris, lichen planus, lichen sclerosus, lupus miliaris disseminatus faciei, melasma, moles, molluscum contagiosum, nail growth rate, nail health, neurodermatitis, nummular eczema, occupational acne, oil acne, onychomycosis, physical urticara, pilonidal cyst, pityriasis rosea, pityriasis versicolor, poison ivy, pomade acne, pseudofolliculitis barbae or acne keloidalis nuchae, psoriasis, psoriatic arthritis, pressure or delayed pressue urticara, puncture wounds such as cuts and scrapes, rash, rare or water type urticara, rhinoplasty, ringworm, rosacea, rothmund-thomson syndrome, sagging of the skin, scabis, scars, seborrhea, seborrheic dermatitis, shingles, skin cancer, skin tag, solar type urticara, spider bite, stretch marks, sunburn, tar acne, tropical acne, thinning of skin, thrush, tinea versicolor, transient acantholytic dermatosis, tycoon's cap or acne necrotica miliaris, uneven skin tone, varicose veins, venous eczema, vibratory angioedema, vitiligo, warts, Weber-Christian disease, wrinkles, x-linked hypohidrotic ectodermal dysplasia, xerotic eczema, yeast infection and general signs of aging.
  • In some embodiments, there is provided methods of treating, controlling or preventing dry skin with the present compositions. In some embodiments profilaggrin (a protein which is converted to filaggrin) is a protein of interest (e.g. when treating ichthyosis vulgaris).
  • In some embodiments, there is provided methods of treating, controlling or preventing any one of the various types of psoriasis (e.g. plague psoriasis, guttate psoriasis, pustular psoriasis, inverse psoriasis, and erythrodermic psoriasis). In various embodiments, the protein of interest is any of the products of the genes psoriasis susceptibility 1 through 9 (PSORSI-PSORS9).
  • Various embodiments relate to the treatment, control, or prevention of eczema (e.g. atopic dermatitis, nummular eczema, dyshidrotic eczema, seborrheic dermatitis, irritant contact dermatitis, allergic contact dermatitis, dyshidrosis, venous eczema, dermatitis herpetiformis, neurodermatitis, autoeczematization and xerotic eczema) and, optionally, one or more of the following may be targeted: filaggrin; three genetic variants, ovo-like 1 (OVOL1), actin-like 9 (ACTL9) and kinesin family member 3 A (KIF3A) have been associated with eczema; and the genes brain-derived neurotrophic factor (BDNF) and tachykinin, precursor 1 (TAC1).
  • Hives, or urticaria, including, but not limited to, acute urticaria, chronic urticara and angioedema, physical urticara, pressure or delayed pressue urticara, cholinergic or stress uricara, cold type urticara, heat type urticara, solar type urticara, rare or water type urticara, vibratory angioedema, exercise-induced anaphylasis and dermatographism may be treated with the present compositions by, for example, targeting PLCG-2.
  • Various embodiments relate to the treatment, control, or prevention of rosacea, which includes, but is not limited to, erthematotelangiectatic rosacea, papulopustular rosacea, phymatous rosacea, and ocular rosacea. Optionally, cathelicidin antimicrobial peptide (CAMP) and/or kallikrein-related peptidase 5 (also known as stratum corneum tryptic enzyme (SCTE)) are proteins of interest.
  • In some embodiments, there is provided methods of treating, controlling or preventing acne with the present compositions. For example, acne may include, but is not limited to, acneiform eruptions, acne aestivalis, acne conglobata, acne cosmetic, acne fulminans, acne keloidalis nuchae, acne mechanica, acne medicamentosa, acne miliaris necrotica, acne necrotica, acne rosacea, baby acne, blackheads, chloracne, excoriated acne, halogen acne, infantile acne or neonatal acne, lupus miliaris disseminatus faciei, occupational acne, oil acne, pomade acne, tar acne, tropical acne, tycoon's cap or acne necrotica miliaris, pseudofolliculitis barbae or acne keloidalis nuchae, and hidradenitis suppurativa. In these embodiments, the protein of interest may be one or more matrix metalloproteinases (MMP), e.g., matrix metalloproteinase-1 (MMP-1 or interstitial collagenase), matrix metalloproteinase-9 (MMP-9), and matrix metalloproteinase-13 (MMP-13).
  • In further embodiments, vitiligo is treated with the present compositions, e.g. wherein the NLR family, pyrin domain containing 1 gene (NALP1) gene is targeted.
  • In some embodiments, the present compositions find use in the treatment, control, or prevention of hyprohidrotic ectodermal dysplasia (HED), e.g. via the ectodysplasin A gene (EDA), receptor (EDAR), and receptor associated death domain (EDARADD).
  • In some embodiments, the present compositions find use in the treatment, control, or prevention of balding, or hair thinning (e.g. male pattern baldness, or androgenetic alopecia (AGA)) and, optionally, one or more of the following may be the protein of interest: androgen receptor (AR), ectodysplasin A2 receptor (EDA2R) and lysophosphatidic acid receptor 6 (P2RY5).
  • The present compositions also find use in methods of treatment, control, or prevention of scars and stretch marks (striae), e.g. via collagen, ribosomal s6 kinase, sectrected phosphoprotein 1 (also known as osteopontin), or transforming growth factor beta 3.
  • Epidermodysplasia verruciformis (also known as Lutz-Lewandowsky epidermodysplasia), a rare autosomal recessive genetic hereditary skin disorder, may also be treated with compositions of the present invention, e.g. by targeted transmembrane channel-like 6 (EVER1) or transmembrane channellike 8 (EVER2) genes.
  • In some embodiments, skin sagging, thinning or wrinkling may be treated, controlled or prevented with present composition, e.g. by targeting one or more of the proteins of interest such as collagen, elastin, fibroblast growth factor 7, TIMP metallopeptidase inhibitors, matrix metallopeptidases, superoxide dismutase and other extracellular matrix proteins and proteoglycans.
  • Further embodiments are used in tanning of the skin, such as via melanocyte-stimulating hormone and/or pro-opiomelanocortin.
  • In some embodiments, the present compositions may be used for wound treatment. In some embodiments, methods of treating, controlling or preventing wounds with the present compositions comprises additional steps of, for example, cleaning the wound bed to facilitate wound healing and closure, including, but not limited to: debridement, sharp debridement (surgical removal of dead or infected tissue from a wound), optionally including chemical debriding agents, such as enzymes, to remove necrotic tissue; wound dressings to provide the wound with a moist, warm environment and to promote tissue repair and healing (e.g., wound dressings comprising hydrogels (e.g., AQUASORB; DUODERM), hydrocolloids (e.g., AQUACEL; COMFEEL), foams (e.g., LYOFOAM; SPYROSORB), and alginates (e.g., ALGISITE; CURASORB); administration of growth factors to stimulate cell division and proliferation and to promote wound healing e.g. becaplermin; and (iv) soft-tissue wound coverage, a skin graft may be necessary to obtain coverage of clean, non-healing wounds (e.g., autologous skin grafts, cadaveric skin graft, bioengineered skin substitutes (e.g., APLIGRAF; DERMAGRAFT)).
  • In various embodiments, the nucleic acid drug described herein can be used in a variety of cosmetic/plastic surgery procedures, including, without limitation, a surgical procedure involving skin grafting and an aesthetic or cosmetic surgery (e.g. a facial plastic surgery procedure including, but not limited to blepharoplasty, rhinoplasty, rhytidectomy, genioplasty, facial implants, otoplasty, hair implantation, cleft lip and cleft palate repair, and/or a body plastic surgery procedure including but not limited to abdominoplasty, brachioplasty, thigh lift, breast reduction, breast augmentation, body contouring, liposuction, hand surgery).
  • In various embodiments, a variety of cancers are treated, controlled or prevented with the present compositions (e.g., colorectal cancer, gallbladder cancer, lung cancer, pancreatic cancer, and stomach cancer). In some embodiments, skin cancer is treated with the present compositions. For instance, the skin cancer is one or more of actinic keratosis, basal cell carcinoma, melanoma, Kaposi's sarcoma, and squamous cell carcinoma. In some embodiments, the present compositions are used adjuvant to complete circumferential peripheral and deep margin assessment, Mohs surgery, radiation (e.g. external beam radiotherapy or brachytherapy), chemotherapy (including but not limited to topical chemotherapy, e.g. with imiquimod or 5-fluorouracil), and cryotherapy. The present compositions also find use in the treatment of various stages of cancers, including skin cancers (e.g. basal cell cancer (BCC), squamous cell cancer (SCC), and melanoma), such as a stage of the American Joint Committee on Cancer (AJCC) TNM system (e.g. one or more of TX, T0, Tis, T1, T1a, T1b, T2, T2A, T2B, T3, T3a, T3b, T4, T4a, T4b, NX, N0, N1, N2, N3, M0, M1a, M1b, M1c) and/or a staging system (e.g. Stage 0, Stage IA, Stage IB, Stage IIA, Stage IIB, Stage IIC, Stage IIIA, Stage IIIB, Stage IIIC, Stage IV).
  • Illustrative cancers and/or tumors of the present invention include, but are not limited to, a basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulval cancer; lymphoma including Hodgkin's and non-Hodgkin's lymphoma, as well as B-cell lymphoma (including low grade/follicular non-Hodgkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; and Waldenstrom's Macroglobulinemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); Hairy cell leukemia; chronic myeloblastic leukemia; as well as other carcinomas and sarcomas; and post-transplant lymphoproliferative disorder (PTLD), as well as abnormal vascular proliferation associated with phakomatoses, edema (such as that associated with brain tumors), and Meigs' syndrome.
  • In various embodiments, one or more rare diseases are treated, controlled or prevented with the present compositions, including, by way of illustration, Erythropoietic Protoporphyria, Hailey-Hailey Disease, Epidermolysis Bullosa (EB), Xeroderma Pigmentosum, Ehlers-Danlos Syndrome, Cutis Laxa, Protein C & Protein S Deficiency, Alport Syndrome, Striate Palmoplantar Keratoderma, Lethal Acantholytic EB, Pseudoxanthoma Elasticum (PXE), Ichthyosis Vulgaris, Pemphigus Vulgaris, and Basal Cell Nevus Syndrome.
  • In various embodiments, the present compositions are used to treat, control or prevent one or more inflammatory diseases or conditions, such as inflammation, acute inflammation, chronic inflammation, respiratory disease, atherosclerosis, restenosis, asthma, allergic rhinitis, atopic dermatitis, septic shock, rheumatoid arthritis, inflammatory bowel disease, inflammatory pelvic disease, pain, ocular inflammatory disease, celiac disease, Leigh Syndrome, Glycerol Kinase Deficiency, Familial eosinophilia (FE), autosomal recessive spastic ataxia, laryngeal inflammatory disease; Tuberculosis, Chronic cholecystitis, Bronchiectasis, Silicosis and other pneumoconioses.
  • In various embodiments, the present compositions are used to treat, control or prevent one or more autoimmune diseases or conditions, such as multiple sclerosis, diabetes mellitus, lupus, celiac disease, Crohn's disease, ulcerative colitis, Guillain-Barre syndrome, scleroderms, Goodpasture's syndrome, Wegener's granulomatosis, autoimmune epilepsy, Rasmussen's encephalitis, Primary biliary sclerosis, Sclerosing cholangitis, Autoimmune hepatitis, Addison's disease, Hashimoto's thyroiditis, Fibromyalgia, Menier's syndrome; transplantation rejection (e.g., prevention of allograft rejection) pernicious anemia, rheumatoid arthritis, systemic lupus erythematosus, dermatomyositis, Sjogren's syndrome, lupus erythematosus, multiple sclerosis, myasthenia gravis, Reiter's syndrome, Grave's disease, and other autoimmune diseases.
  • In various embodiments, the present compositions are used to treat, control or prevent one or more neurologic diseases, including ADHD, AIDS—Neurological Complications, Absence of the Septum Pellucidum, Acquired Epileptiform Aphasia, Acute Disseminated Encephalomyelitis, Adrenoleukodystrophy, Agenesis of the Corpus Callosum, Agnosia, Aicardi Syndrome, Alexander Disease, Alpers' Disease, Alternating Hemiplegia, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Anencephaly, Aneurysm, Angelman Syndrome, Angiomatosis, Anoxia, Aphasia, Apraxia, Arachnoid Cysts, Arachnoiditis, Arnold-Chiari Malformation, Arteriovenous Malformation, Aspartame, Asperger Syndrome, Ataxia Telangiectasia, Ataxia, Attention Deficit-Hyperactivity Disorder, Autism, Autonomic Dysfunction, Back Pain, Barth Syndrome, Batten Disease, Behcet's Disease, Bell's Palsy, Benign Essential Blepharospasm, Benign Focal Amyotrophy, Benign Intracranial Hypertension, Bernhardt-Roth Syndrome, Binswanger's Disease, Blepharospasm, Bloch-Sulzberger Syndrome, Brachial Plexus Birth Injuries, Brachial Plexus Injuries, Bradbury-Eggleston Syndrome, Brain Aneurysm, Brain Injury, Brain and Spinal Tumors, Brown-Sequard Syndrome, Bulbospinal Muscular Atrophy, Canavan Disease, Carpal Tunnel Syndrome, Causalgia, Cavernomas, Cavernous Angioma, Cavernous Malformation, Central Cervical Cord Syndrome, Central Cord Syndrome, Central Pain Syndrome, Cephalic Disorders, Cerebellar Degeneration, Cerebellar Hypoplasia, Cerebral Aneurysm, Cerebral Arteriosclerosis, Cerebral Atrophy, Cerebral Beriberi, Cerebral Gigantism, Cerebral Hypoxia, Cerebral Palsy, Cerebro-Oculo-Facio-Skeletal Syndrome, Charcot-Marie-Tooth Disorder, Chiari Malformation, Chorea, Choreoacanthocytosis, Chronic Inflammatory Demyelinating Polyneuropathy (CIDP), Chronic Orthostatic Intolerance, Chronic Pain, Cockayne Syndrome Type II, Coffin Lowry Syndrome, Coma, including Persistent Vegetative State, Complex Regional Pain Syndrome, Congenital Facial Diplegia, Congenital Myasthenia, Congenital Myopathy, Congenital Vascular Cavernous Malformations, Corticobasal Degeneration, Cranial Arteritis, Craniosynostosis, Creutzfeldt-Jakob Disease, Cumulative Trauma Disorders, Cushing's Syndrome, Cytomegalic Inclusion Body Disease (CIBD), Cytomegalovirus Infection, Dancing Eyes-Dancing Feet Syndrome, Dandy-Walker Syndrome, Dawson Disease, De Morsier's Syndrome, Dejerine-Klumpke Palsy, Dementia—Multi-Infarct, Dementia—Subcortical, Dementia With Lewy Bodies, Dermatomyositis, Developmental Dyspraxia, Devic's Syndrome, Diabetic Neuropathy, Diffuse Sclerosis, Dravet's Syndrome, Dysautonomia, Dysgraphia, Dyslexia, Dysphagia, Dyspraxia, Dystonias, Early Infantile Epileptic Encephalopathy, Empty Sella Syndrome, Encephalitis Lethargica, Encephalitis and Meningitis, Encephaloceles, Encephalopathy, Encephalotrigeminal Angiomatosis, Epilepsy, Erb's Palsy, Erb-Duchenne and Dejerine-Klumpke Palsies, Fabry's Disease, Fahr's Syndrome, Fainting, Familial Dysautonomia, Familial Hemangioma, Familial Idiopathic Basal Ganglia Calcification, Familial Spastic Paralysis, Febrile Seizures (e.g., GEFS and GEFS plus), Fisher Syndrome, Floppy Infant Syndrome, Friedreich's Ataxia, Gaucher's Disease, Gerstmann's Syndrome, Gerstmann-Straussler-Scheinker Disease, Giant Cell Arteritis, Giant Cell Inclusion Disease, Globoid Cell Leukodystrophy, Glossopharyngeal Neuralgia, Guillain-Barre Syndrome, HTLV-1 Associated Myelopathy, Hallervorden-Spatz Disease, Head Injury, Headache, Hemicrania Continua, Hemifacial Spasm, Hemiplegia Alterans, Hereditary Neuropathies, Hereditary Spastic Paraplegia, Heredopathia Atactica Polyneuritiformis, Herpes Zoster Oticus, Herpes Zoster, Hirayama Syndrome, Holoprosencephaly, Huntington's Disease, Hydranencephaly, Hydrocephalus—Normal Pressure, Hydrocephalus, Hydromyelia, Hypercortisolism, Hypersomnia, Hypertonia, Hypotonia, Hypoxia, Immune-Mediated Encephalomyelitis, Inclusion Body Myositis, Incontinentia Pigmenti, Infantile Hypotonia, Infantile Phytanic Acid Storage Disease, Infantile Refsum Disease, Infantile Spasms, Inflammatory Myopathy, Intestinal Lipodystrophy, Intracranial Cysts, Intracranial Hypertension, Isaac's Syndrome, Joubert Syndrome, Kearns-Sayre Syndrome, Kennedy's Disease, Kinsbourne syndrome, Kleine-Levin syndrome, Klippel Feil Syndrome, Klippel-Trenaunay Syndrome (KTS), Klüver-Bucy Syndrome, Korsakoffs Amnesic Syndrome, Krabbe Disease, Kugelberg-Welander Disease, Kuru, Lambert-Eaton Myasthenic Syndrome, Landau-Kleffner Syndrome, Lateral Femoral Cutaneous Nerve Entrapment, Lateral Medullary Syndrome, Learning Disabilities, Leigh's Disease, Lennox-Gastaut Syndrome, Lesch-Nyhan Syndrome, Leukodystrophy, Levine-Critchley Syndrome, Lewy Body Dementia, Lissencephaly, Locked-In Syndrome, Lou Gehrig's Disease, Lupus—Neurological Sequelae, Lyme Disease—Neurological Complications, Machado-Joseph Disease, Macrencephaly, Megalencephaly, Melkersson-Rosenthal Syndrome, Meningitis, Menkes Disease, Meralgia Paresthetica, Metachromatic Leukodystrophy, Microcephaly, Migraine, Miller Fisher Syndrome, Mini-Strokes, Mitochondrial Myopathies, Mobius Syndrome, Monomelic Amyotrophy, Motor Neuron Diseases, Moyamoya Disease, Mucolipidoses, Mucopolysaccharidoses, Multi-Infarct Dementia, Multifocal Motor Neuropathy, Multiple Sclerosis, Multiple System Atrophy with Orthostatic Hypotension, Multiple System Atrophy, Muscular Dystrophy, Myasthenia—Congenital, Myasthenia Gravis, Myelinoclastic Diffuse Sclerosis, Myoclonic Encephalopathy of Infants, Myoclonus, Myopathy—Congenital, Myopathy—Thyrotoxic, Myopathy, Myotonia Congenita, Myotonia, Narcolepsy, Neuroacanthocytosis, Neurodegeneration with Brain Iron Accumulation, Neurofibromatosis, Neuroleptic Malignant Syndrome, Neurological Complications of AIDS, Neurological Manifestations of Pompe Disease, Neuromyelitis Optica, Neuromyotonia, Neuronal Ceroid Lipofuscinosis, Neuronal Migration Disorders, Neuropathy—Hereditary, Neurosarcoidosis, Neurotoxicity, Nevus Cavernosus, Niemann-Pick Disease, O'Sullivan-McLeod Syndrome, Occipital Neuralgia, Occult Spinal Dysraphism Sequence, Ohtahara Syndrome, Olivopontocerebellar Atrophy, Opsoclonus Myoclonus, Orthostatic Hypotension, Overuse Syndrome, Pain—Chronic, Paraneoplastic Syndromes, Paresthesia, Parkinson's Disease, Parnyotonia Congenita, Paroxysmal Choreoathetosis, Paroxysmal Hemicrania, Parry-Romberg, Pelizaeus-Merzbacher Disease, Pena Shokeir II Syndrome, Perineural Cysts, Periodic Paralyses, Peripheral Neuropathy, Periventricular Leukomalacia, Persistent Vegetative State, Pervasive Developmental Disorders, Phytanic Acid Storage Disease, Pick's Disease, Piriformis Syndrome, Pituitary Tumors, Polymyositis, Pompe Disease, Porencephaly, Post-Polio Syndrome, Postherpetic Neuralgia, Postinfectious Encephalomyelitis, Postural Hypotension, Postural Orthostatic Tachycardia Syndrome, Postural Tachycardia Syndrome, Primary Lateral Sclerosis, Prion Diseases, Progressive Hemifacial Atrophy, Progressive Locomotor Ataxia, Progressive Multifocal Leukoencephalopathy, Progressive Sclerosing Poliodystrophy, Progressive Supranuclear Palsy, Pseudotumor Cerebri, Pyridoxine Dependent and Pyridoxine Responsive Siezure Disorders, Ramsay Hunt Syndrome Type I, Ramsay Hunt Syndrome Type II, Rasmussen's Encephalitis and other autoimmune epilepsies, Reflex Sympathetic Dystrophy Syndrome, Refsum Disease—Infantile, Refsum Disease, Repetitive Motion Disorders, Repetitive Stress Injuries, Restless Legs Syndrome, Retrovirus-Associated Myelopathy, Rett Syndrome, Reye's Syndrome, Riley-Day Syndrome, SUNCT Headache, Sacral Nerve Root Cysts, Saint Vitus Dance, Salivary Gland Disease, Sandhoff Disease, Schilder's Disease, Schizencephaly, Seizure Disorders, Septo-Optic Dysplasia, Severe Myoclonic Epilepsy of Infancy (SMEI), Shaken Baby Syndrome, Shingles, Shy-Drager Syndrome, Sjogren's Syndrome, Sleep Apnea, Sleeping Sickness, Soto's Syndrome, Spasticity, Spina Bifida, Spinal Cord Infarction, Spinal Cord Injury, Spinal Cord Tumors, Spinal Muscular Atrophy, Spinocerebellar Atrophy, Steele-Richardson-Olszewski Syndrome, Stiff-Person Syndrome, Striatonigral Degeneration, Stroke, Sturge-Weber Syndrome, Subacute Sclerosing Panencephalitis, Subcortical Arteriosclerotic Encephalopathy, Swallowing Disorders, Sydenham Chorea, Syncope, Syphilitic Spinal Sclerosis, Syringohydromyelia, Syringomyelia, Systemic Lupus Erythematosus, Tabes Dorsalis, Tardive Dyskinesia, Tarlov Cysts, Tay-Sachs Disease, Temporal Arteritis, Tethered Spinal Cord Syndrome, Thomsen Disease, Thoracic Outlet Syndrome, Thyrotoxic Myopathy, Tic Douloureux, Todd's Paralysis, Tourette Syndrome, Transient Ischemic Attack, Transmissible Spongiform Encephalopathies, Transverse Myelitis, Traumatic Brain Injury, Tremor, Trigeminal Neuralgia, Tropical Spastic Paraparesis, Tuberous Sclerosis, Vascular Erectile Tumor, Vasculitis including Temporal Arteritis, Von Economo's Disease, Von Hippel-Lindau disease (VHL), Von Recklinghausen's Disease, Wallenberg's Syndrome, Werdnig-Hoffman Disease, Wernicke-Korsakoff Syndrome, West Syndrome, Whipple's Disease, Williams Syndrome, Wilson's Disease, X-Linked Spinal and Bulbar Muscular Atrophy, and Zellweger Syndrome.
  • In various embodiments, the present compositions are used to treat one or more respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), bronchiectasis, allergic rhinitis, sinusitis, pulmonary vasoconstriction, inflammation, allergies, impeded respiration, respiratory distress syndrome, cystic fibrosis, pulmonary hypertension, pulmonary vasoconstriction, emphysema, Hantavirus pulmonary syndrome (HPS), Loeffler's syndrome, Goodpasture's syndrome, Pleurisy, pneumonitis, pulmonary edema, pulmonary fibrosis, Sarcoidosis, complications associated with respiratory syncitial virus infection, and other respiratory diseases.
  • In various embodiments, the present compositions are used to treat, control or prevent cardiovascular disease, such as a disease or condition affecting the heart and vasculature, including but not limited to, coronary heart disease (CHD), cerebrovascular disease (CVD), aortic stenosis, peripheral vascular disease, atherosclerosis, arteriosclerosis, myocardial infarction (heart attack), cerebrovascular diseases (stroke), transient ischaemic attacks (TIA), angina (stable and unstable), atrial fibrillation, arrhythmia, vavular disease, and/or congestive heart failure.
  • In various embodiments, the present compositions are used to treat, control or prevent one or more metabolic-related disorders. In various embodiments, the present invention is useful for the treatment, controlling or prevention of diabetes, including Type 1 and Type 2 diabetes and diabetes associated with obesity. The compositions and methods of the present invention are useful for the treatment or prevention of diabetes-related disorders, including without limitation diabetic nephropathy, hyperglycemia, impaired glucose tolerance, insulin resistance, obesity, lipid disorders, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels, high LDL levels, atherosclerosis and its sequelae, vascular restenosis, irritable bowel syndrome, inflammatory bowel disease, including Crohn's disease and ulcerative colitis, other inflammatory conditions, pancreatitis, abdominal obesity, neurodegenerative disease, retinopathy, neoplastic conditions, adipose cell tumors, adipose cell carcinomas, such as liposarcoma, prostate cancer and other cancers, including gastric, breast, bladder and colon cancers, angiogenesis, Alzheimer's disease, psoriasis, high blood pressure, Metabolic Syndrome (e.g. a person has three or more of the following disorders: abdominal obesity, hypertriglyceridemia, low HDL cholesterol, high blood pressure, and high fasting plasma glucose), ovarian hyperandrogenism (polycystic ovary syndrome), and other disorders where insulin resistance is a component, such as sleep apnea. The compositions and methods of the present invention are useful for the treatment, control, or prevention of obesity, including genetic or environmental, and obesity-related disorders. The obesity-related disorders herein are associated with, caused by, or result from obesity. Examples of obesity-related disorders include obesity, diabetes, overeating, binge eating, and bulimia, hypertension, elevated plasma insulin concentrations and insulin resistance, dyslipidemia, hyperlipidemia, endometrial, breast, prostate, kidney and colon cancer, osteoarthritis, obstructive sleep apnea, gallstones, heart disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovary disease, craniopharyngioma, Prader-Willi Syndrome, Frohlich's syndrome, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia. Further examples of obesity-related disorders are Metabolic Syndrome, insulin resistance syndrome, reproductive hormone abnormalities, sexual and reproductive dysfunction, such as impaired fertility, infertility, hypogonadism in males and hirsutism in females, fetal defects associated with maternal obesity, gastrointestinal motility disorders, such as obesity-related gastro-esophageal reflux, respiratory disorders, such as obesity-hypoventilation syndrome (Pickwickian syndrome), breathlessness, cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, lower back pain, gallbladder disease, hyperuricemia, gout, and kidney cancer, and increased anesthetic risk. The compositions and methods of the present invention are also useful to treat Alzheimer's disease.
  • Nucleic acids, including liposomal formulations containing nucleic acids, when delivered in vivo, can accumulate in the liver and/or spleen. It has now been discovered that nucleic acids encoding proteins can modulate protein expression in the liver and spleen, and that nucleic acids used in this manner can constitute potent therapeutics for the treatment of liver and spleen diseases. Certain embodiments are therefore directed to a method for treating liver and/or spleen disease by delivering to a patient a nucleic acid encoding a protein of interest. Other embodiments are directed to a therapeutic composition comprising a nucleic acid encoding a protein of interest, for the treatment of liver and/or spleen disease. Diseases and conditions of the liver and/or spleen that can be treated include, but are not limited to: hepatitis, alcohol-induced liver disease, drug-induced liver disease, Epstein Barr virus infection, adenovirus infection, cytomegalovirus infection, toxoplasmosis, Rocky Mountain spotted fever, non-alcoholic fatty liver disease, hemochromatosis, Wilson's Disease, Gilbert's Disease, and cancer of the liver and/or spleen.
  • In some embodiments, the present compositions and methods relate to the treatment of type 1 diabetes, heart disease, including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, cystic fibrosis, sickle-cell anemia, thalassemia, Fanconi anemia, severe combined immunodeficiency, hereditary sensory neuropathy, xeroderma pigmentosum, Huntington's disease, muscular dystrophy, amyotrophic lateral sclerosis, Alzheimer's disease, cancer, and infectious diseases including hepatitis and HIV/AIDS.
  • Further, in some embodiments, the present methods and compositions find use in targeting any of the proteins or in treatment of any of the diseases or disorders of Table 2B. In various embodiments, the present invention contemplates the targeting of the full-length and/or truncated forms of any of the proteins disclosed in Table 2B. In various embodiments, the present invention contemplates the targeting of the precursor forms and/or mature forms and/or isoforms of any of the proteins disclosed in Table 2B.
  • In various embodiments, the present invention contemplates the targeting of a protein having about 60% (e.g. about 60%, or about 61%, or about 62%, or about 63%, or about 64%, or about 65%, or about 66%, or about 67%, or about 68%, or about 69%, or about 70%, or about 71%, or about 72%, or about 73%, or about 74%, or about 75%, or about 76%, or about 77%, or about 78%, or about 79%, or about 80%, or about 81%, or about 82%, or about 83%, or about 84%, or about 85%, or about 86%, or about 87%, or about 88%, or about 89%, or about 90%, or about 91%, or about 92%, or about 93%, or about 94%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99%) sequence identity with any of the protein sequences disclosed herein (e.g. in Table 2B).
  • In various embodiments, the present invention contemplates the targeting of a protein comprising an amino acid sequence having one or more amino acid mutations relative to any of the protein sequences described herein (e.g. in Table 2B). For example, the present invention contemplates the targeting of a protein comprising an amino acid sequence having 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12 amino acid mutations relative to any of the protein sequences described herein (e.g. in Table 2B). In some embodiments, the one or more amino acid mutations may be independently selected from substitutions, insertions, deletions, and truncations.
  • In some embodiments, the amino acid mutations are amino acid substitutions, and may include conservative and/or non-conservative substitutions. “Conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved. The 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1) hydrophobic: Met, Ala, Val, Leu, Ile; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gln; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe.
  • As used herein, “conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide. In addition, glycine and proline may be substituted for one another based on their ability to disrupt α-helices.
  • As used herein, “non-conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1) to (6) shown above.
  • In various embodiments, the substitutions may also include non-classical amino acids (e.g. selenocysteine, pyrrolysine, N-formylmethionine β-alanine, GABA and δ-Aminolevulinic acid, 4-aminobenzoic acid (PABA), D-isomers of the common amino acids, 2,4-diaminobutyric acid, α-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, γ-Abu, ε-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosme, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β methyl amino acids, C α-methyl amino acids, N α-methyl amino acids, and amino acid analogs in general).
  • In Table 2B, all Illustrative Identifiers (e.g. Gene Seq nos. and references are hereby incorporated by reference in their entireties).
  • TABLE 2B
    Illustrative Proteins, Illustrative Peptides, and Illustrative Indications
    Protein/Peptide
    Illustrative Identifier
    Reference Description/Illustrative Indication(s)
    Transthyretin (TTR) his gene encodes transthyretin, one of the three prealbumins including alpha-1-
    (SEQ ID NOs: 637 and 638) antitrypsin, transthyretin and orosomucoid. Transthyretin is a carrier protein; it
    Gene ID: 7276 transports thyroid hormones in the plasma and cerebrospinal fluid, and also
    transports retinol (vitamin A) in the plasma. The protein consists of a tetramer of
    identical subunits. More than 80 different mutations in this gene have been reported;
    most mutations are related to anyloid deposition, affecting predominantly peripheral
    nerve and/or the heart, and a small portion of the gene mutations is non-
    amyloidogenic. The diseases caused by mutations include amyloidotic polyneuropathy,
    euthyroid hyperthyroxinaemia, amyloidotic vitreous opacities, cardiomyopathy,
    oculoleptomeningeal amyloidosis, meningocerebrovascular amyloidosis, carpal tunnel
    syndrome, etc.
    Endothelial Cell Specific This gene encodes a secreted protein which is mainly expressed in the endothelial
    Molecule
    1 cells in human lung and kidney tissues. The expression of this gene is regulated by
    (SEQ ID NO: 629 to 632) cytokines, suggesting that it may play a role in endothelium-dependent pathological
    Gene ID: 11082 disorders. The transcript contains multiple polyadenylation and mRNA instability
    signals. Two transcript variants encoding different isoforms have been found for this gene
    Parathyroid hormone PTH is secreted by the chief cells of the parathyroid glands. PTH plays an important
    P01270|PTHY_HUMAN role in the regulation of serum calcium, serum phosphate, and vitamin D synthesis.
    Parathyroid hormone
    (SEQ ID NO: 508)
    BMP-1 GeneSeq BMP1 belongs to the transforming growth factor-beta (TGFB) super- family. Bone
    Accession P80618 morphogenic proteins induce cartilage and bone formation, play important role in
    WO8800205 nephrogesis, and play an important role in the development of many organs,
    P13497/BMP1_HUMAN including lung, heart, teeth, gut, skin, and particularly the kidney. BMP-1 activity can
    Bone morphogenetic be determined using the following assays known in the art: Nat Genet. 2001 January;
    protein 1 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274,
    (isoform BMP1-3) Issue 16, 10897-10902, Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10,
    (SEQ ID NO: 169) 1580-1594. Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    P13497-2|BMP1_HUMAN
    Isoform BMP1-1 of Bone
    morphogenetic protein 1
    (isoform BMP1-1)
    (SEQ ID NO: 509)
    P13497-3|BMP1_HUMAN
    Isoform BMP1-4 of Bone
    morphogenetic protein 1
    (isoform BMP1-4)
    (SEQ ID NO: 510)
    P13497-4|BMP1_HUMAN
    Isoform BMP1-5 of Bone
    morphogenetic protein 1
    (isoform BMP1-5)
    (SEQ ID NO: 511)
    P13497-5|BMP1_HUMAN
    Isoform BMP1-6 of Bone
    morphogenetic protein 1
    (isoform BMP1-6)
    (SEQ ID NO: 512)
    P13497-6|BMP1_HUMAN
    Isoform BMP1-7 of Bone
    morphogenetic protein 1
    (isoform BMP1-7)
    (SEQ ID NO: 513)
    BMP-2 GeneSeq BMP-2 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession P80619 morphogenic protein induces bone formation. BMP-2 activity can be determined
    WO8800205 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    P12643/BMP2_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    protein 2 Induction of Cartilage, Tissue and Bone Growth, and Diabetes. Infarction recovery.
    (SEQ ID NO: 170) Bone repair. Osteoporosis.
    BMP-3 BMP-3 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    P12645|BMP3_HUMAN morphogenic protein induces bone formation. BMP-3 activity can be determined
    Bone morphogenetic using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    protein
    3 Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    (SEQ ID NO: 514) Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    BMP-2B GeneSeq BMP-2b belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession W24850 U.S. morphogenic protein induces bone formation. BMP-2b activity can be determined
    Pat. No. 5,631,142 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    P12644/BMP4_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; I Biol Cbcre, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594. Induction of
    protein 4 Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 171)
    BMP-4 GeneSeq BMP-4 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession B02796 morphogenic protein induces bone formation. BMP-4 activity can be determined
    WO0020591 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    P12644/BMP4_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    protein 4 Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 172)
    BMP-5 GeneSeq BMP-5 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession B02797 morphogenic protein induces bone formation. BMP-5 activity can be determined
    WO0020591 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    P22003/BMP5_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    protein 5 Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (isoform 1)
    (SEQ ID NO: 173)
    P22003-2|BMP5_HUMAN
    Isoform
    2 of Bone
    morphogenetic protein 5
    (isoform 2)
    (SEQ ID NO: 515)
    BMP-6 GeneSeq BMP-6 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession R32904 U.S. morphogenic protein induces bone formation. BMP-6 activity can be determined
    Pat. No. 5,187,076 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    P22004/BMP6_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    protein 6 Induction of Cartilage, Tissue and Bone Growth, and Diabetes. Hemochromatosis.
    (SEQ ID NO: 174)
    Osteogenic Protein-1; OP- OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    1; BMP-7 GeneSeq morphogenic protein induces bone formation. OP-1 activity can be determined using
    Accession W34783 the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    WO973462 Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    P18075/BMP7_HUMAN Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    Bone morphogenetic Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    protein 7
    (SEQ ID NO: 175)
    BMP7 Variant A OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    (SEQ ID NO: 579) morphogenic protein induces bone formation. OP-1 activity can be determined using
    the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    BMP7 Variant B OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    (SEQ ID NO: 580) morphogenic protein induces bone formation. OP-1 activity can be determined using
    the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    BMP7 Variant C OP-1 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    (SEQ ID NO: 581) morphogenic protein induces bone formation. OP-1 activity can be determined using
    the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    Osteogenic Protein-2 OP-2 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    GeneSeq Accession morphogenic protein induces bone formation. OP-2 activity can be determined using
    R57973 WO9406399 the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    P34820/BMP8B_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 0, 1580-1594.
    protein 8B Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 176)
    GDF-1 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to
    Accession R60961 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    WO9406449 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    P27539/GDF1_HUMAN Cell Biol. 4: 172 178; Miyazono, K. et al., (1994) Adv. Immunol. 55: 181-220).
    Embryonic Activation of this heteromeric receptor complex occurs when TGF-beta binds to
    growth/differentiation TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    factor 1 propagates the signal to downstream targets (Chen, F. and Weinberg, R. A. (1995)
    (SEQ ID NO: 177) PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341 347). The effect of
    GDF-1 on signaling can be assayed by treating Primary BAECs transferred with a
    construct called p3TP-Lux, containing a TGF-beta responsive promoter fused to a
    reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature
    370: 341-347). Developmental disorders, Induction of Cartilage, Tissue and Bone
    Growth, and Diabetes
    BMP-9 GeneSeq BMP-9 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession R86903 morphogenic protein induces bone formation. BMP-9 activity can be determined
    WO9533830 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    Q9UK05/GDF2_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Growth/differentiation Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    factor 2 Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 178)
    BMP-10 GeneSeq BMP-10 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession R66202 morphogenic protein induces bone formation. BMP-10 activity can be determined
    WO9426893 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    O95393/BMP10_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    protein 10 Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 179)
    BMP-12 GeneSeq BMP-12 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession R78734 morphogenic protein induces bone formation. BMP-12 activity can be determined
    WO9516035 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    Q7Z4P5/GDF7_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Growth/differentiation Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    factor 7 Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 180)
    BMP-15 GeneSeq BMP-15 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession W11261 morphogenic protein induces bone formation. BMP-15 activity can be determined
    W09636710 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    O95972/BMP15_HUMAN Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Bone morphogenetic Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    protein 15 Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    (SEQ ID NO: 181)
    BMP-17 GeneSeq BMP-17 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession Y17870 morphogenic protein induces bone formation. BMP-17 activity can be determined
    WO9929718 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    SEQ ID NO: 2 from U.S. Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Pat. No. 7,151,086 Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    (SEQ ID NO: 182) Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    BMP-18 GeneSeq BMP-18 belongs to the transforming growth factor-beta (TGFB) superfamily. Bone
    Accession Y17871 morphogenic protein induces bone formation. BMP-18 activity can be determined
    WO9929718 using the following assays known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J
    SEQ ID NO: 4 from U.S. Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902,
    Pat. No. 7,151,086 Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594.
    (SEQ ID NO: 183) Induction of Cartilage, Tissue and Bone Growth, and Diabetes
    Inhibin alpha GeneSeq The inhibin beta A subunit joins the alpha subunit to form a pituitary FSH secretion
    Accession B02806 inhibitor. Inhibin has been shown to regulate gonadal stromal cell proliferation
    WO0020591 negatively and to have tumor-suppressor activity. In addition, serum levels of inhibin
    P05111/INHA_HUMAN have been shown to reflect the size of granulosa-cell tumors and can therefore be
    Inhibin alpha chain used as a marker for primary as well as recurrent disease. Tumor suppressor activity
    (SEQ ID NO: 184) of inhibin can be determined using assays known in the art: Matzuk et al., Nature
    1992 Nov. 26: 360 (6402); 313-9. Tumor suppression.
    Inhibin beta GeneSeq The inhibin beta A subunit joins the alpha subunit to form a pituitary FSH secretion
    Accession H02808 inhibitor. Inhibin has been shown to regulate gonadal stromal cell proliferation
    WO0020591 negatively and to have tumour-suppressor activity. In addition, serum levels of inhibin
    P08476/INHBA_HUMAN have been shown to reflect the size of granulosa-cell tumors and can therefore be
    Inhibin beta A chain used as a marker for primary as well as recurrent disease. Tumor suppressor activity
    (SEQ ID NO: 185) of inhibin can be determined using assays known in the art: Matzuk et al., Nature
    P09529/INHBB_HUMAN 1992 Nov. 26: 360 (6402); 313-9. Tumor suppression.
    Inhibin beta B chain
    (SEQ ID NO: 186)
    Cerberus Protein Cerebus is believed to be involved in the inhibition of BMP activity BMP activity, in the
    GeneSeq Accession presence of the antagonist Cerebus, can be determined using the following assays
    W86032 WO9849296 known in the art: Nat Genet. 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr. 1;
    O95813/CER1_HUMAN 237(1): 295-302; J Biol Chem, Vol. 274, Issue 16, 10897-10902, Apr. 16, 1999; and
    Cerberus Hogan, B. L. M. (1996) Genes Dev. 10, 1580-1594. BMP Antagonist useful for
    (SEQ ID NO: 187) Osteosarcoma, abnormal bone growth.
    Soluble BMP Receptor Soluble BMP receptor kinase protein-3 is involved in the binding of BMPs. Soluble
    Kinase Protein-3 BMP receptor kinase protein-3 is useful as an antagonist for the inhibition of BMP
    GeneSeq Accession activity. BMP activity, in the presence of the soluble antagonist BMP receptor kinase
    R95227 WO9614579 protein-3, can be determined using the following assays known in the art: Nat Genet.
    Q13873/BMPR2_HUMAN 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem,
    Bone morphogenetic Vol. 274, Issue 16, 10897-10902, Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes
    protein receptor type-2 Dev. 10, 1580-1594. BMP Antagonist useful for Osteosarcoma, abnormal bone
    (SEQ ID NO: 188) growth.
    BMP Processing Enzyme BMPs belong to the transforming growth factor-beta (TGFB) superfamily. Bone
    Furin GeneSeq Accession morphogenic protein induces bone formation. BMP activity, in the presence of the
    W36099 WO9741250 Furin, can be determined using the following assays known in the art: Nat Genet.
    P09958/FURIN_HUMAN 2001 January; 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol.
    Furin 274, Issue 16, 10897-10902, Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Dev.
    (SEQ ID NO: 189) 10, 1580-1594. Bone formation or Regeneration Abnormalities
    TGF-beta 1 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to
    Accession R29657 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    WO9216228 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    P01137/TGFB1_HUMAN Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
    Transforming growth Activation of this heteromeric receptor complex occurs when TGF-beta. binds to
    factor beta-1 TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    (SEQ ID NO: 190) propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995)
    PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341. The effect of TGF
    betas on signaling can be assayed by treating Primary BAECs transfected with a
    construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a
    reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature
    370: 341-347). Useful for treating cancer and to promote wound healing.
    TGF-beta 2 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to
    Accession R39659 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    EP542679 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    P61812/TGFB2_HUMAN Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
    Transforming growth Activation of this heteromeric receptor complex occurs when TGF-beta. binds to
    factor beta-2 TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    (SEQ ID NO: 191) propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995)
    PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341. The effect of TGF
    betas on signaling can be assayed by treating Primary BAECs transfected with a
    construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a
    reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature
    370: 341-347). Useful for treating cancer and to promote wound healing.
    ZTGF-beta 9 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to
    Accession Y70654 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    WO0015798 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    SEQ ID NO: 2 of Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
    WO0015798 Activation of this heteromeric receptor complex occurs when TGF-beta. binds to
    (SEQ ID NO: 192) TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995)
    PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341. The effect of TGF
    betas on signaling can be assayed by treating Primary BAECs transfected with a
    construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a
    reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature
    370: 341-347). Useful for treating cancer and to promote wound healing.
    Anti-TGF beta family Members of the TGF-beta family of proteins initiate cell signaling by binding to
    antibodies GB2305921 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
    Activation of this heteromeric receptor complex occurs when TGF-beta. binds to
    TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995)
    PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341. The effect of TGF
    betas on signaling in the presence of an anti-TGF beta antibody, can be assayed by
    treating Primary BAECs transfected with a construct called p3TP-Lux, containing a
    TGF-beta responsive promoter fused to a reporter gene, and measuring luciferase
    gene expression (Wrana et al., 1994, Nature 370: 341-347). Useful for control of
    fibrosis, immune, and inflammatory disease.
    Latent TGF beta binding Members of the TGF-beta family of proteins initiate cell signaling by binding to
    protein II GeneSeq heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    Accession Y70552 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    WO0012551 Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
    Q14767/LTBP2_HUMAN Activation of this heteromeric receptor complex occurs when TGF-beta. binds to
    Latent-transforming TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    growth factor beta-binding propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995)
    protein 2 PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341. The effect of TGF
    (SEQ ID NO: 193) betas on signaling in the presence of a TGF beta binding protein, can be assayed by
    treating Primary BAECs transfected with a construct called p3TP-Lux, containing a
    TGF-beta responsivepromoter fused to a reporter gene, and measuring luciferase
    gene expression (Wrana et al., 1994, Nature 370: 341-347). Useful for inhibiting
    tissue or tumor growth.
    MP52 GeneSeq Members of the TGF-beta family of proteins initiate cell signaling by binding to
    Accession W36100 heteromeric receptor complexes of type I (TbetaRI) and type II (TbetaRII)
    WO9741250 serine/threonine kinase receptors (reviewed by Massague, J. et al. (1994) Trends
    P43026/GDF5_HUMAN Cell Biol. 4: 172 178; Miyazono, K. et al. (1994) Adv. Immunol. 55: 181-220).
    Growth/differentiation Activation of this heteromeric receptor complex occurs when TGF-beta. binds to
    factor 5 TbetaRII, which then recruits and phosphorylates TbetaRI. Activated TbetaRI then
    (SEQ ID NO: 194) propagates the signal to downstream targets (Chen, F. and Weinberg. R. A. (1995)
    PNA892: 1565-1569; Wrana, J. L. et al. (1994) Nature 370: 341. The effect of TGF
    betas on signaling can be assayed by treating Primary BAECs transfected with a
    construct called p3TP-Lux, containing a TGF- beta responsive promoter fused to a
    reporter gene, and measuring luciferase gene expression (Wrana et al., 1994, Nature
    370: 341-347). Bone formation or Regeneration Abnormalities
    b57 Protein GeneSeq BMPs are involved in the induction of bone formation. Specific antagonists are useful
    Accession W69293 is preventing this activity from occurring. BMP activity, in the presence of b57 protein,
    WO9837195 can be determined using the following assays known in the art: Nat Genet. 2001 January;
    SEQ ID NO: 2 of 27(1): 84-8; Eur J Biochem 1996 Apr. 1; 237(1): 295-302; J Biol Chem, Vol. 274, Issue
    WO9837195
    16, 1089-10902, Apr. 16, 1999; and Hogan, B. L. M. (1996) Genes Deve. 10, 1580-1594.
    (SEQ ID NO: 195) BMP Antagonist useful for Osteosarcoma, abnormal bone growth.
    Resistin GeneSeq This gene belongs to the family defined by mouse FIZZI and FIZZ3/Resistin genes.
    Accession W69293 The characteristic feature of this family is the C-terminal stretch of 10 cys residues
    WO0064920 with identical spacing. The mouse homolog of this protein is secreted by adipocytes,
    Q9HD89/RETN_HUMAN may be the hormone potentially linking obesity to type II diabetes. Ability of resistin to
    Resistin influence type II diabetes can be determined using assays known in the art: Pontoglio
    (isoform 1) et al., J Clin Invest 1998 May 15; 101(10): 2215-22. Type II diabetes and Syndrome
    (SEQ ID NO: 196) X.
    Galectin-4 GeneSeq Galectins are a family of carbohydrate-binding proteins characterized by an affinity for
    Accession W11841 beta- galactoside containing glycoconjugates. Ability of Galectin-4 polypeptides to
    WO9703190 bind lactose can be determined using assays known in the art: Wada, et al., J Biol
    P56470/LEG4_HUMAN Chem 1997 Feb. 28; 272(9): 6078-86. Lactose intolerance.
    Galectin-4
    (SEQ ID NO: 197)
    APM-I; ACRP- 30; ACPR30 gene is exclusively expressed in adipose tissue. ACRP30 is thought to
    Famoxin GeneSeq increase fatty acid oxidation by muscle tissue. Ability of ACRP30 polypeptides to
    Accession Y71035 influence obesity and fat oxidation can be determined using assays known in the art:
    W00026363 Fruebis et al., Proc Nat'l Acad Sci USA 2001 Feb. 13; 98(4): 2005-10. Obesity,
    Q15848/ADIPO_HUMAN Metabolic disorders, Lipid Metabolism; Hormone Secretion.
    Adiponectin
    (SEQ ID NO: 198)
    ACRP-30 Homologue; ACPR30 gene is exclusively expressed in adipose tissue. ACRP30 is thought to
    Complement Component increase fatty acid oxidation by muscle tissue. Ability of ACRP30 homologue
    Clq C GeneSeq Accession polypeptides to influence obesity and fat oxidation can be determined using assays
    B30234 WO0063376 known in the art: Fruebis et al., Proc Nat'l Acad Sci USA 2001 Feb. 13; 98(4): 2005-10.
    P02747/C1QC_HUMAN Obesity, Metabolic disorders, Lipid Metabolism; Hormone Secretion.
    Complement C1q
    subcomponent subunit C
    (SEQ ID NO: 199)
    Calpain-10a GeneSeq Calpain is believed to play a role in insulin secretion and insulin activity, and therefore
    Accession Y79567 may be useful in the treatment of type II diabetes. Ability of Calpain-10 to influence
    WO0023603 type II diabetes can be determined using assays known in the art: Pontoglio et al., J
    Q9HC96/CAN10_HUMAN Clin Invest 1998 May 15; 101(10): 2215-22. Diabetes mellitus; Regulation of Insulin
    Calpain-10 secretory response; Insulin mediated glucose transport disorders.
    (Isoform A)
    (SEQ ID NO: 200)
    Calpain-10b GeneSeq Calpain is believed to play a role in insulin secretion and insulin activity, and therefore
    Accession Y79568 may be useful in the treatment of type II diabetes. Ability of Calpain-10 to influence
    WO0023603 type II diabetes can be determined using assays known in the art: Pontoglio et al., J
    Q9HC96- Clin Invest 1998 May 15; 101(10): 2215-22. Diabetes mellitus; Regulation of Insulin
    2/CAN10_HUMAN Isoform secretory response; Insulin mediated glucose transport disorders.
    B of Calpain-10
    (SEQ ID NO: 201)
    Calpain-10c GeneSeq Calpain is believed to play a role in insulin secretion and insulin activity, and therefore
    Accession Y79569 may be useful in the treatment of type II diabetes. Ability of Calpain-10 to influence
    WO0023603 type II diabetes can be determined using assays known in the art: Pontoglio et al., J
    Q9HC96- Clin Invest 1998 May 15; 101(10): 2215-22. Diabetes mellitus; Regulation of Insulin
    3/CAN10_HUMAN Isoform secretory response; Insulin mediated glucose transport disorders.
    C of Calpain-10
    (SEQ ID NO: 202)
    PDGF-D GeneSeq Vascular Endothelial Growth Factor. Proliferation assay using NR6R-3T3 cells
    Accession Y71130 (Rizzino 1988 Cancer Res. 48: 4266). Wound Healing; Atherosclermis.
    WO0027879
    Q9GZP0/PDGFD_HUMAN
    Platelet-derived growth
    factor D
    (isoform 1)
    (SEQ ID NO: 203)
    FasL GeneSeq Accession Activities associated with apoptosis and immune system functions. Activity can be
    Y28594 WO9936079 determined using Apoptosis assays known in the art: Walczak et al. (1996) EMBOJ
    P48023/TNFL6_HUMAN 16: 5386-5397. Apoptosis-related disorders; Auto- immune disorders; Graft v-Host
    Tumor necrosis factor disorders.
    ligand superfamily
    member 6
    (isoform 1)
    (SEQ ID NO: 204)
    Chondro modulin-like Chondromodulin proteins are cartilage proteins thought to confer resistance to
    protein GeneSeq anglogeneis, and thus are useful as anti-angiogenic agents that may have utility in
    Accession Y71262 combating cancer. Ability of Chondromodulin-like protein to inhibit vascularization can
    WO0029579 be determined using assays known in the art: Hirakie et al., J Biol Chem 1997 Dec.
    SEQ ID NO: 2 from 19; 272(51): 32419-26. Antianglogenic agent; Osteoblast proliferation stimulator;
    WO0029579 prevents vascularization of cartilage tissue; Useful to treat cancer.
    (SEQ ID NO: 370)
    Patched GeneSeq Patched is a tumour-suppressor receptor for Sonic hedgehog (shh), which is a protein
    Accession W72969 U.S. that controls developmental patterning and growth. Ability of soluble Patched to bind
    Pat. No. 5,837,538 to and inhibit the activities of shh can be determined using assays known in the art:
    Q13635/PTC1_HUMAN Stone et al., Nature 1996 Nov. 14; 384(6605): 129-34. Receptor for Hedgehog cellular
    Protein patched homolog 1 proliferation signaling molecule. This receptor is useful as a means of preventing
    (isoform L) cellular proliferation via the shh signaling path- way, thus useful for cancers.
    (SEQ ID NO: 205)
    Patched-2 GeneSeq Patched is a tumour-suppressor receptor for Sonic hedgehog (shh), which is a protein
    Accession Y43261 that controls developmental patterning and growth. Ability of soluble Patched to bind
    WO9953058 to and inhibit the activities of shh can be determined using assays known in the art:
    Q9Y6C5/PTC2_HUMAN Stone et al., Nature 1996 Nov. 14; 384(6605): 129-34. Receptor for Hedgehog cellular
    Protein patched homolog 2 proliferation signaling molecule. This receptor is useful as a means of preventing
    (isoform 1) cellular proliferation via the shh signaling path- way, thus useful for cancers.
    (SEQ ID NO: 206)
    Maspin; Protease Inhibitor Maspin is a member of the serpin family of serine protease inhibitors that is thought to
    5 GeneSeq Accession suppress tumor metastasis. The inhibitory effects of Maspin and other protease
    R50938 WO9405804 inhibitors can be assayed using methods known in the art such as a labeled protease
    P36952/SPB5_HUMAN substrate, for example, Universal Protease Substrate (casein, resorufin-labeled):
    Serpin B5 Roche Molecular Biochemicals, Cat. No. 1080733. Tumor suppressor which is down-
    (isoform 1) regulated in breast cancers. The maspin protein has tumour suppressing and
    (SEQ ID NO: 207) invasion sup- pressing activity.
    Endostatin GeneSeq Endostatin is believed to inhibit effects of capillary endothelial cell proliferation. The
    Accession B28399 inhibitory effects of endostatin can be assayed using assays disclosed by Cao et al.
    WO0064946 (1996) J. Biol. Chem. 271 29461-29467. Anti-angiogenic activity. Useful in the
    P39060/COIA1_HUMAN prevention and/or treatment of cancers.
    Collagen alpha-1(XVIII)
    chain
    (isoform 1)
    (SEQ ID NO: 208)
    aFGF; FGF-1 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession P94037 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    EP298723 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    P05230/FGF1_HUMAN be useful as anti-cancer agents. Diabetes, Metabolic Disease, Obesity.
    Fibroblast growth factor 1
    (isoform 1)
    (SEQ ID NO: 209)
    bFGF; FGF-2 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession R06685 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    FR2642086 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    P09038/FGF2_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 2
    (isoform 1)
    (SEQ ID NO: 210)
    FGF-3; INT-2 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession R07824 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    WO9503831 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    P11487/FGF3_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 3
    (SEQ ID NO: 211)
    FGF-4; HST-1; HBGF-4 Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    GeneSeq Accession Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    R07825 WO9503831 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    P08620/FGF4_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 4
    (isoform 1)
    (SEQ ID NO: 212)
    FGF-5 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession W22600 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    WO9730155 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    P12034/FGF5_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 5
    (isoform long)
    (SEQ ID NO: 213)
    FGF-6; Heparin binding Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    secreted transforming Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    factor-2 GeneSeq and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    Accession R58555 be useful as anti-cancer agents.
    EP613946
    P10767/FGF6_HUMAN
    Fibroblast growth factor 6
    (SEQ ID NO: 214)
    FGF-8 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession R80783 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    WO9524928 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    P55075/FGF8_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 8
    (isoform 8E)
    (SEQ ID NO: 215)
    FGF-9; Gila activating Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    factor GeneSeq Accession Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    R70822 WO9503831 and proliferation of cells, such as epithelial cells and keratinocytes. Hair growth.
    P31371/FGF9_HUMAN Antagonists may be useful as anti-cancer agents.
    Fibroblast growth factor 9
    (SEQ ID NO: 216)
    FGF-12; Fibroblast growth Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    factor homologous factor-1 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    GeneSeq Accession and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    W06309 WO9635708 be useful as anti-cancer agents.
    P61328/FGF12_HUMAN
    Fibroblast growth factor 12
    (isoform 1)
    (SEQ ID NO: 217)
    FGF-19 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession Y08582 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    WO9927100 and proliferation of cells, such as epithelial cells and keratinocytes. Chronic liver
    O95750/FGF19_HUMAN disease. Primary biliary cirrhosis. Bile acid-induced liver damage. Antagonists may be
    Fibroblast growth factor 19 useful as anti-cancer agents.
    (SEQ ID NO: 218)
    FGF-16 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession Y05474 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    WO9918128 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    O43320/FGF16_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 16
    (SEQ ID NO: 219)
    FGF-18 GeneSeq Fibroblast Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    Accession Y08590 Cancer Res. 48: 4266); Examples 23 and 39 disclosed herein. Promotion of growth
    WO9927100 and proliferation of cells, such as epithelial cells and keratinocytes. Antagonists may
    O76093/FGF18_HUMAN be useful as anti-cancer agents.
    Fibroblast growth factor 18
    (SEQ ID NO: 220)
    flt-3 ligand GeneSeq Stem Cell Progenitor Chemokine activities can be determined using assays known in
    Accession R67541 the art: Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited
    EP627487 by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa,
    P49771|FLT3L_HUMAN NJ. Promotion of immune cell growth and/or differentiation.
    Fms-related tyrosine
    kinase
    3 ligand
    (isoform 1)
    (SEQ ID NO: 221)
    VEGF-110 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y69417 determined using assays known in the art, such as those disclosed in International
    WO0013702 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 11 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO0013702 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 222)
    VEGF-121 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession B50432 determined using assays known in the art, such as those disclosed in International
    WO0071713 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 2 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO0071713 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 223)
    VEGF-138 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y43483 determined using assays known in the art, such as those disclosed in International
    WO9940197 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 4 of cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO99/40197 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 371)
    VEGF-145 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y69413 determined using assays known in the art, such as those disclosed in International
    WO0013702 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 4 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO0013702 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 224)
    VEGF-162 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y43484 determined using assays known in the art, such as those disclosed in International
    W09940197 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 8 of cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO99/40197 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 372)
    VEGF-165 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y69414 determined using assays known in the art, such as those disclosed in International
    WO0013702 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 6 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO0013702 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 225)
    VEGF-182 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y43483 determined using assays known in the art, such as those disclosed in International
    W09940197 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 6 of cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO99/40197 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 373)
    VEGF-189 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y69415 determined using assays known in the art, such as those disclosed in International
    WO0013702 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 8 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO0013702 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 226)
    VEGF-206 GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession Y69416 determined using assays known in the art, such as those disclosed in International
    W00013702 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 10 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO0013702 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 227)
    VEGF-D GeneSeq Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    Accession W53240 determined using assays known in the art, such as those disclosed in International
    WO9807832 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    O43915/VEGFD_HUMAN cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    Vascular endothelial angiogenic agents, and may be applicable for cancer.
    growth factor D
    (SEQ ID NO: 374)
    VEGF-E; VEGF-X Promotes the growth and/or proliferation of endothelial cells. VEGF activity can be
    GeneSeq Accession determined using assays known in the art, such as those disclosed in International
    Y33679 WO9947677 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    SEQ ID NO: 2 from cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    WO9947677 angiogenic agents, and may be applicable for cancer.
    (SEQ ID NO: 228)
    VEGF Receptor; KDR; flk- Receptor for VEGF polypeptides VEGF activity, in the presence of flk-1 polypeptides,
    1 GeneSeq Accession can be determined using assays known in the art, such as those disclosed in
    W69679 WO9831794 International Publication No. WO0045835, for example. VEGF Receptor. Fusion
    P35968/VGFR2_HUMAN protein with the extracellular domain is useful as an anti-angiogenic agent.
    Vascular endothelial Antagonists may be useful in the promotion of angiogenesis.
    growth factor receptor 2
    (isoform 1)
    (SEQ ID NO: 229)
    Soluble VEGF Receptor Receptor for VEGF polypeptides VEGF activity, in the presence of VEGF Receptor
    GeneSeq Accession polypeptides, can be determined using assays known in the art, such as those
    W47037 U.S. Pat. No. disclosed in International Publication No. WO0045835, for example. VEGF Receptor.
    5,712,380 Fusion protein with the extracellular domain is useful as an anti-angiogenic agent.
    sVEGF-RI (FIG. 3) of Antagonists may be useful in the promotion of angiogenesis.
    U.S. Pat. No. 5,712,380
    (SEQ ID NO: 442)
    sVEGF-RII (FIG. 11) of
    U.S. Pat. No. 5,712,380
    (SEQ ID NO: 443)
    sVEGF-RTMI (FIG. 15)
    of U.S. Pat. No. 5,712,380
    (SEQ ID NO: 444)
    sVEGF-RTMII (FIG. 13)
    of U.S. Pat. No. 5,712,380
    (SEQ ID NO: 445)
    flt-1 GeneSeq Accession Receptor for VEGF polypeptides VEGF activity, in the presence of flt-1 polypeptides,
    Y70751 WO0021560 can be determined using assays known in the art, such as those disclosed in
    P17948/VGFR1_HUMAN International Publication No. WO0045835, for example. VEGF Receptor. Fusion
    Vascular endothelial protein with the extracellular domain is useful as an anti-angiogenic agent.
    growth factor receptor 1 Antagonists may be useful in the promotion of angiogenesis.
    (isoform 1)
    (SEQ ID NO: 230)
    VEGF R-3; flt-4 GeneSeq Receptor for VEGF polypeptides VEGF activity, in the presence of flt-4 polypeptides,
    Accession B29047 can be determined using assays known in the art, such as those disclosed in
    WO0058511 International Publication No. WO0045835, for example. VEGF Receptor. Fusion
    P35916/VGFR3_HUMAN protein with the extracellular domain is useful as an anti-angiogenic agent.
    Vascular endothelial Antagonists may be useful in the promotion of angiogenesis.
    growth factor receptor 3
    (isoform 1)
    (SEQ ID NO: 231)
    Neuropilin-1 GeneSeq Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    Accession Y06319 known in the art, such as those disclosed in International Publication No.
    WO9929858 WO0045835, for example. Promotion of growth and proliferation of cells, such as
    O14786/NRP1_HUMAN vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    Neuropilin-1 may be applicable for cancer.
    (isoform 1)
    (SEQ ID NO: 232)
    Neuropilin-2 GeneSeq Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    Accession Y03618 known in the art, such as those disclosed in International Publication No.
    WO9929858 WO0045835, for example. Promotion of growth and proliferation of cells, such as
    O60462/NRP2_HUMAN vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    Neuropilin-2 may be applicable for cancer.
    (isoform A22)
    (SEQ ID NO: 233)
    Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    muscle troponin C may contribute to the difficulty encountered in revascularizing the ischemic
    GeneSeq Accession myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    W22597 W09730085 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    P02585/TNNC2_HUMAN USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    Troponin C, skeletal
    muscle
    (SEQ ID NO: 234)
    Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    muscle troponin I may contribute to the difficulty encountered in revascularizing the ischemic
    GeneSeq Accession myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    W18054 W09730085 angiogenesis can be determined using assays known in the art. Proc Natl Acad Sci
    P48788/TNNI2_HUMAN USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    Troponin I, fast skeletal
    muscle
    (isoform 1)
    (SEQ ID NO: 235)
    Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    muscle troponin T may contribute to the difficulty encountered in revascularizing the ischemic
    GeneSeq Accession myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    W22599 W09730085 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    SEQ ID NO: 3 of USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    WO9730085
    (SEQ ID NO: 236)
    fragment. myofibrillar Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    protein troponin I may contribute to the difficulty encountered in revascularizing the ischemic
    GeneSeq Accession myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    W18053 W09719955 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    SEQ ID NO: 3 of USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    WO9719955
    (SEQ ID NO: 237)
    myofibrillar protein Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    troponin I GeneSeq may contribute to the difficulty encountered in revascularizing the ischemic
    Accession W18054 myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    WO9719955 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    SEQ ID NO: 3 of USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    WO9719955
    (SEQ ID NO: 237)
    Troponin peptides Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    GeneSeq Accessions may contribute to the difficulty encountered in revascularizing the ischemic
    Y29581, Y29582, Y29583, myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    Y29584, Y29585, and angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    Y29586 WO9933874 USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    Wildtype troponins
    provided as:
    Human fast twitch skeletal
    muscle troponin C
    GeneSeq Accession
    W22597 W09730085
    P02585/TNNC2_HUMAN
    Troponin C, skeletal
    muscle
    (SEQ ID NO: 234)
    Human fast twitch skeletal
    muscle troponin I
    GeneSeq Accession
    W18054 W09730085
    P48788/TNNI2_HUMAN
    Troponin I, fast skeletal
    muscle
    (isoform 1)
    (SEQ ID NO: 235)
    Human fast twitch skeletal
    muscle troponin T
    GeneSeq Accession
    W22599 W09730085
    SEQ ID NO: 3 of
    WO9730085
    (SEQ ID NO: 236)
    fragment. myofibrillar
    protein troponin I
    GeneSeq Accession
    W18053 W09719955
    SEQ ID NO: 3 of
    WO9719955
    (SEQ ID NO: 237)
    Human fast twitch skeletal
    muscle Troponin subunit C
    GeneSeq Accession
    B00134 WO0054770
    SEQ ID NO: 1 of
    WO0054770
    (SEQ ID NO: 375)
    Human fast twitch skeletal
    muscle Troponin subunit I
    Protein GeneSeq
    Accession B00135
    WO0054770
    SEQ ID NO: 2 of
    WO0054770
    (SEQ ID NO: 376)
    Human fast twitch skeletal
    muscle Troponin subunit T
    GeneSeq Accession
    B00136 WO0054770
    SEQ ID NO: 3 of
    WO0054770
    (SEQ ID NO: 377)
    Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    muscle Troponin subunit C may contribute to the difficulty encountered in revascularizing the ischemic
    GeneSeq Accession myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    B00134 WO0054770 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    SEQ ID NO: 1 of USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    WO0054770
    (SEQ ID NO: 375)
    Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    muscle Troponin subunit I may contribute to the difficulty encountered in revascularizing the ischemic
    Protein GeneSeq myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    Accession B00135 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    WO0054770 USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    SEQ ID NO: 2 of
    WO0054770
    (SEQ ID NO: 376)
    Human fast twitch skeletal Troponins are contractile proteins that are thought to inhibit angiogenesis. High levels
    muscle Troponin subunit T may contribute to the difficulty encountered in revascularizing the ischemic
    GeneSeq Accession myocardium after cardiovascular injury. Ability of soluble Troponins to inhibit
    B00136 WO0054770 angiogenesis can be determined using assays known in the art:. Proc Natl Acad Sci
    SEQ ID NO: 3 of USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis
    WO0054770
    (SEQ ID NO: 377)
    Activator Inbibitor-1; PAI-1 PAIs are believed to play a role in cancer, and cardiovascular disease and blood-
    GeneSeq Accession clotting disorders. Methods that measure plasminogen activator inhibitor (PAI) activity
    R08411 WO9013648 are known in the art, for example, assay the ability of PAI to inhibit tissue
    P05121/PAI1_HUMAN plasminogen activator (tPA) or urokinase (uPA): J Biochem Biophys Methods 2000
    Plasminogen activator Sep. 11; 45(2): 127-40, Breast Cancer Res Treat 1996; 41(2): 141-6. Methods that
    inhibitor 1 measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad
    (isoform 1) Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders.
    (SEQ ID NO: 238)
    Plasminogen Activator PAIs are believed to play a role in cancer, and cardiovascular disease and blood-
    Inhibitor-2; PAI-2 clotting disorders. Methods that measure plasminogen activator inhibitor (PAI) activity
    GeneSeq Accession are known in the art, for example, assay the ability of PAI to inhibit tissue
    P94160 DE3722673 plasminogen activator (tPA) or urokinase (uPA): J Biochem Biophys Methods 2000
    P05120/PAI2_HUMAN Sep. 11; 45(2): 127-40, Breast Cancer Res Treat 1996; 41(2): 141-6. Methods that
    Plasminogen activator measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad
    inhibitor
    2 Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders.
    (SEQ ID NO: 239)
    Activator Inhibitor-2; PAI-2 PAIs are believed to play a role in cancer, and cardiovascular disease and blood-
    GeneSeq Accession clotting disorders. Methods that measure plasminogen activator inhibitor (PAI) activity
    R10921 WO9102057 are known in the art, for example, assay the ability of PAI to inhibit tissue
    P05120/PAI2_HUMAN plasminogen activator (tPA) or urokinase (uPA): J Biochem Biophys Methods 2000
    Plasminogen activator Sep. 11; 45(2): 127-40, Breast Cancer Res Treat 1996; 41(2): 141-6. Methods that
    inhibitor 2 measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad
    (SEQ ID NO: 239) Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders.
    Human PAI-1 mutants PAIs are believed to play a role in cancer, and cardiovascular disease and blood-
    GeneSeq Accessions clotting disorders. Methods that measure plasminogen activator inhibitor (PAI) activity
    R11755, R11756, R11757, are known in the art, for example, assay the ability of PAI to inhibit tissue
    R11758, R11759, R11760, plasminogen activator (tPA) or urokinase (uPA): J Biochem Biophys Methods 2000
    R11761, R11762 and Sep. 11; 45(2): 127-40, Breast Cancer Res Treat 1996; 41(2): 141-6. Methods that
    R11763 WO9105048 measure anti- angiogenesis activity are known in the art, for example, Proc Natl Acad
    Wildtype PAI-1 is provided Sci USA 1999 Mar. 16; 96(6): 2645-50. Anti-angiogenesis; blood-clotting disorders.
    as P05121/PAI1_HUMAN
    Plasminogen activator
    inhibitor 1
    (isoform 1)
    (SEQ ID NO: 238)
    CXCR3; CXC GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession Y79372 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO0018431 Members of this family are involved in a similarly diverse range of pathologies
    P49682|CXCR3_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-X-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G-
    type 3 protein coupled receptors. Over 40 human chemokines have been described, which
    (isoform 1) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 240) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Soluble CXCR3 polypeptides may be useful for
    inhibiting chemokine activities and viral infection.
    Modified Rantes GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W38129 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9737005 Members of this family are involved in a similarly diverse range of pathologies
    Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    herein as chemokines exert their effects by acting on a family of seven transmembrane G-
    P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 5 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    RANTES GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession Y05299 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    EP905240 Members of this family are involved in a similarly diverse range of pathologies
    P13501/CCL5_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 5 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 241) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    MCP-Ia GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession R73914 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9509232 Members of this family are involved in a similarly diverse range of pathologies
    MCP-1 provided as including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P13500/CCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 2 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 337) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    MCP-Ib GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession Y26176 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9929728 Members of this family are involved in a similarly diverse range of pathologies
    MCP-1 provided as including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P13500/CCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 2 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 337) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    MCP-I receptor GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession R79165 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9519436 Members of this family are involved in a similarly diverse range of pathologies
    MCP-IA including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9519436 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 446) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    MCP-1B using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    SEQ ID NO: 4 of Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    WO9519436 Humana Press Inc., Totowa, NJ. Soluble MCP-1 Receptor polypep- tides may be
    (SEQ ID NO: 447) useful for inhibiting chemokine activities and viral infection.
    MCP-3 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession R73915 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W09509232 Members of this family are involved in a similarly diverse range of pathologies
    P80098/CCL7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 7 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 336) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    MCP-4 receptor GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W56689 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9809171 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9809171 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 378) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Soluble MCP-4 Receptor polypep- tides may be
    useful for inhibiting chemokine activities and viral infection.
    RANTES receptor Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W29588 U.S. Pat. No. Members of this family are involved in a similarly diverse range of pathologies
    5,652,133 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of U.S. Pat. chemokines exert their effects by acting on a family of seven transmembrane G-
    No. 5,652,133 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 379) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Soluble RANTES Receptor polypep- tides may be
    useful for inhibiting chemokine activities and viral infection.
    CCR5 variant GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W88238 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9854317 Members of this family are involved in a similarly diverse range of pathologies
    Variants of wildtype CCR5 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    which has the sequence chemokines exert their effects by acting on a family of seven transmembrane G-
    of: protein coupled receptors. Over 40 human chemokines have been described, which
    P51681|CCR5_HUMAN bind to ~17 receptors thus far identified. Chemokine activities can be determined
    C-C chemokine receptor using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    type 5 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 448) Humana Press Inc., Totowa, NJ. Soluble CCR5 polypeptides may be useful for
    inhibiting chemokine activities and viral infection.
    CCR7 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession B50859 U.S. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Pat. No. 6,153,441 Members of this family are involved in a similarly diverse range of pathologies
    P32248/CCR7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G-
    type 7 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 243) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Soluble CCR7 polypeptides may be useful for
    inhibiting chemokine activities and viral infection.
    CXC3 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W23345 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9727299 Members of this family are involved in a similarly diverse range of pathologies
    P78423/X3CL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Fractalkine chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 244) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    Eotaxin GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W10099 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9700960 Members of this family are involved in a similarly diverse range of pathologies
    P51671/CCL11_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Eotaxin chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 245) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    Neurotactin GeneSeq Neurotactin may play a role in chemotactic leukocyte migration and brain
    Accessions Y77537, inflammation processes. Chemotactic leukocyte migration assays are known in the
    W34307, Y53259, and, art, for example: J. Immunol. Methods 33, ((1980)); Nature 1997 Jun. 5; 387(6633):
    Y77539 U.S. Pat. No. 611-7. Immune disorders.
    6,013,257 WO9742224
    P78423/X3CL1_HUMAN
    Fractalkine
    (SEQ ID NO: 244)
    Human CKbeta-9 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B50860 U.S. Pat. No. Members of this family are involved in a similarly diverse range of pathologies
    6,153,441 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of U.S. Pat. chemokines exert their effects by acting on a family of seven transmembrane G-
    No. 6,153,441 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 246) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    Lymphotactin GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession B50052 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO0073320 Members of this family are involved in a similarly diverse range of pathologies
    P47992/XCL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Lymphotactin chemokines exert their effects by acting on a family of seven transmembrane G.
    (SEQ ID NO: 247) Chemokine activities can be determined using assays known in the art: Methods in
    Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot,
    T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    Immune disorders.
    MIP-3 alpha GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W44398 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9801557 Members of this family are involved in a similarly diverse range of pathologies
    P78556/CCL20_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 20 chemokines exert their effects by acting on a family of seven transmembrane G.
    (isoform 1) Chemokine activities can be determined using assays known in the art: Methods in
    (SEQ ID NO: 248) Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot,
    T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    Immune disorders.
    MIP-3 beta GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W44399 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9801557 Members of this family are involved in a similarly diverse range of pathologies
    Q99731/CCL19_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 19 chemokines exert their effects by acting on a family of seven transmembrane G.
    (SEQ ID NO: 249) Chemokine activities can be determined using assays known in the art: Methods in
    Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot,
    T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    Immune disorders.
    MIP-Gamma GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession R70798 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO2006135382 Members of this family are involved in a similarly diverse range of pathologies
    (SEQ ID NO: 457) including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    chemokines exert their effects by acting on a family of seven transmembrane G.
    Chemokine activities can be determined using assays known in the art: Methods in
    Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot,
    T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    Immune disorders.
    Stem Cell Inhibitory Factor Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    R11553 WO9104274 Members of this family are involved in a similarly diverse range of pathologies
    SCIF in Table I of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9104274 chemokines exert their effects by acting on a family of seven transmembrane G.
    (SEQ ID NO: 380) Chemokine activities can be determined using assays known in the art: Methods in
    SCIF in Table II of Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot,
    WO9104274 T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    (SEQ ID NO: 381) Hematopoietic growth factors.
    Thrombopoietin GeneSeq Thrombopoietin is involved in the regulation of the growth and differentiation of
    Accession R79905 megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    WO9521920 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    P40225|TPO_HUMAN 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Thrombopoietin Hematopoietic growth factors.
    (isoform 1)
    (SEQ ID NO: 250)
    c-kit ligand; SCF; Mast cell C-kit ligan is thought to stimulate the proliferation of mast cells, and is able to
    growth factor; MGF; augment the proliferation of both myeloid and lymphoid hematopoietic progenitors in
    Fibrosarcoma- derived bone marrow culture. C-kit ligand is also though to act synergistically with other
    stem cell factor GeneSeq cytokines. Chemokine activities can be determined using assays known in the art:
    Accession Y53284, Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot,
    R83978 and R83977 T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    EP992579 and EP676470 Hematopoietic growth factors.
    P21583|SCF_HUMAN Kit
    ligand
    (isoform 1)
    (SEQ ID NO: 251)
    Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    factor GeneSeq Accession known in the art, such as those disclosed in International Publication No.
    B48653 WO0066736 WO0045835, for example. Promotion of growth and proliferation of cells, such as
    PDGF-A vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    P04085/PDGFA_HUMAN may be applicable for cancer.
    Platelet-derived growth
    factor subunit A
    (Isoform long)
    (SEQ ID NO: 257)
    PDGF-B
    P01127/PDGFB_HUMAN
    Platelet-derived growth
    factor subunit B
    (isoform 1)
    (SEQ ID NO: 258)
    Melanoma inhibiting Melanoma inhibiting protein has melanoma-inhibiting activity and can be used to treat
    protein GeneSeq cancer (melanoma, glioblastoma, neuroblastoma, small cell lung cancer,
    Accession R69811 neuroectodermal tumors) or as an immunosuppressant (it inhibits IL-2 or
    WO9503328 phytohaemagglutinin induced proliferation of peripheral blood lymphocytes. Tumor
    (SEQ ID NO: 458) suppressor activity of melanoma inhibiting protein can be determined using assays
    known in the art: Matzuk et al., Nature 1992 Nov. 26; 360(6402): 313-9. Cancer;
    melanoma
    Glioma-derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    factor GeneSeq Accession known in the art, such as those disclosed in International Publication No.
    R08120 EP399816 WO0045835, for example. Promotion of growth and proliferation of cells, such as
    vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    may be applicable for cancer.
    Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    factor precursor A known in the art, such as those disclosed in International Publication No.
    GeneSeq Accession WO0045835, for example. Promotion of growth and proliferation of cells, such as
    R84759 EP682110 vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    PDGF-A precursor (variant may be applicable for cancer.
    D1)
    (SEQ ID NO: 382)
    PDGF-A precursor (variant
    13-1)
    (SEQ ID NO: 383)
    Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    factor precursor B known in the art, such as those disclosed in International Publication No.
    GeneSeq Accession WO0045835, for example. Promotion of growth and proliferation of cells, such as
    R84760 EP682110, FIG. vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    1 or FIG. 2 may be applicable for cancer.
    Wildtype PDGF-B
    provided as:
    PDGF-B
    P01127/PDGFB_HUMAN
    Platelet-derived growth
    factor subunit B
    (isoform 1)
    (SEQ ID NO: 258)
    Platelet derived growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    factor Bvsis GeneSeq known in the art, such as those disclosed in International Publication No.
    Accession P80595 and WO0045835, for example. Promotion of growth and proliferation of cells, such as
    P80596 EP282317 vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    FIG. 1 of EP282317 may be applicable for cancer.
    (SEQ ID NO: 384)
    Placental Growth Factor Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    GeneSeq Accessions known in the art, such as those disclosed in International Publication No.
    R23059 and R23060 WO0045835, for example. Promotion of growth and proliferation of cells, such as
    WO9206194 vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    P49763-2/PLGF_HUMAN may be applicable for cancer.
    Isoform PIGF-1 of
    Placenta growth factor
    (isoform PIGF-1)
    (SEQ ID NO: 252)
    Placental Growth Factor-2 Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    GeneSeq Accession known in the art, such as those disclosed in International Publication No.
    Y08289 DE19748734 WO0045835, for example. Promotion of growth and proliferation of cells, such as
    P49763-3/PLGF_HUMAN vascular endothelial cells. Antagonists may be useful as anti-angiogenic agents, and
    Isoform PIGF-2 of may be applicable for cancer.
    Placenta growth factor
    (isoform PIGF-2)
    (SEQ ID NO: 253)
    Thrombopoietin Thrombopoietin is involved in the regulation of the growth and differentiation of
    derivative1 GeneSeq megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Accession Y77244 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Thrombopoietin Thrombopoietin is involved in the regulation of the growth and differentiation of
    derivative2 GeneSeq megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Accession Y77255 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Thrombopoietin derivative Thrombopoietin is involved in the regulation of the growth and differentiation of
    3 GeneSeq Accession megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Y77262 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Thrombopoietin derivative Thrombopoietin is involved in the regulation of the growth and differentiation of
    4 GeneSeq Accession megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Y77267 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Thrombopoietin derivative Thrombopoietin is involved in the regulation of the growth and differentiation of
    5 GeneSeq Accession megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Y77246 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Thrombopoietin derivative Thrombopoietin is involved in the regulation of the growth and differentiation of
    6 GeneSeq Accession megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Y77253 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Thrombopoietin derivative Thrombopoietin is involved in the regulation of the growth and differentiation of
    7 GeneSeq Accession megakaryocytes and preceptors thereof. Thrombopoietin (TPO) can be assayed to
    Y77256 determine regulation of growth and differentiation of megakaryocytes. Mol Cell Biol
    WO0000612 (e.g. Table 3) 2001 April; 21(8): 2659-70; Exp Hematol 2001 January; 29(1): 51-8 and within.
    Wildtype thrombopoietin Thrombocytopenia, cancer.
    provided as:
    P40225|TPO_HUMAN
    Thrombopoietin
    (isoform 1)
    (SEQ ID NO: 250)
    Fractalkine GeneSeq Fractalkine is believed to play a role in chemotactic leukocyte migration and
    Accession Y53255 U.S. neurological disorders. Fractalkine activity can be determined using Chemotactic
    Pat. No. 6,043,086 leukocyte migration assays known in the art, for example: J. Immunol. Methods 33,
    P78423/X3CL1_HUMAN ((1980)); Nature 1997 Jun. 5; 387(6633): 611-7. Immune disorders.
    Fractalkine
    (SEQ ID NO: 244)
    CXC3 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W23345 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9757599 Members of this family are involved in a similarly diverse range of pathologies
    P78423/X3CL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Fractalkine chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 244) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders.
    CCR7 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession B50859 U.S. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Pat. No. 6,153,441 Members of this family are involved in a similarly diverse range of pathologies
    P32248/CCR7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G-
    type 7 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 243) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Soluble CCR7 polypeptides may be useful for
    inhibiting chemokine activities and viral infection.
    Nerve Growth Factor-beta Nerve Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988 Cancer
    GeneSeq Accession Res. 48: 4266) Neurological disorders, cancer
    R11474 EP414151
    P01138/NGF_HUMAN
    Beta-nerve growth factor
    (SEQ ID NO: 254)
    Nerve Growth Factor- Nerve Growth Factor Proliferation assay using NR6R 3T3 cells (Rizzino 1988 Cancer
    beta2 GeneSeq Accession Res. 48: 4266 Neurological disorders, cancer
    W69725 EP859056
    FIG. 1 of EP859056
    (SEQ ID NO: 465)
    Neurotrophin-3 GeneSeq Neurotrophins regulate neuronal cell survival and synaptic plasticity. Trk tyrosine
    Accession W8889 kinase activation assays known in the art can be used to assay for neurotrophin
    WO9821234 activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
    P20783/NTF3_HUMAN Neurological disorders, cancer
    Neurotrophin-3
    (isoform 1)
    (SEQ ID NO: 255)
    Neurotrophin-4 GeneSeq Neurotrophins regulate neuronal cell survival and synaptic plasticity. Trk tyrosine
    Accession R47100 kinase activation assays known in the art can be used to assay for neurotrophin
    WO9325684 activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
    P34130/NTF4_HUMAN Neurological disorders, cancer
    Neurotrophin-4
    (SEQ ID NO: 256)
    Neurotrophin- 4a Neurotrophins regulate neuronal cell survival and synaptic plasticity. Trk tyrosine
    GeneSeq Accession kinase activation assays known in the art can be used to assay for neurotrophin
    R47101 WO9325684 activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
    Wildtype neurotrophin Neurological disorders, cancer
    provided as:
    P34130/NTF4_HUMAN
    Neurotrophin-4
    (SEQ ID NO: 256)
    Neurotrophin- 4b Neurotrophins regulate neuronal cell survival and synaptic plasticity. tyrosine kinases.
    GeneSeq Accession Trk tyrosine kinase activation assays known in the art can be used to assay for
    R47102 WO9325684 neurotrophin activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
    P34130/NTF4_HUMAN Neurological disorders, cancer
    Neurotrophin-4
    (SEQ ID NO: 256)
    Neurotrophin- 4c Neurotrophins regulate neuronal cell survival and synaptic plasticity. tyrosine kinases.
    GeneSeq Accession Trk tyrosine kinase activation assays known in the art can be used to assay for
    R47103 WO9325684 neurotrophin activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
    P34130/NTF4_HUMAN Neurological disorders, cancer
    Neurotrophin-4
    (SEQ ID NO: 256)
    Neurotrophin- 4d Neurotrophins regulate neuronal cell survival and synaptic plasticity. tyrosine kinases.
    GeneSeq Accession Trk tyrosine kinase activation assays known in the art can be used to assay for
    R47102 WO9325684 neurotrophin activity, for example, Proc Natl Acad Sci USA 2001 Mar. 13; 98(6): 3555-3560.
    P34130/NTF4_HUMAN Neurological disorders, cancer
    Neurotrophin-4
    (SEQ ID NO: 256)
    Platelet-Derived Growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    Factor A chain GeneSeq known in the art, such as those disclosed in International Publication No.
    Accession R38918 U.S. W00045835, for example. Promotion of growth and proliferation of cells, such as
    Pat. No. 5,219,739 vascular endothelial cells. Hematopoietic and immune dis- orders. Antagonists may
    P04085/PDGFA_HUMAN be useful as anti-angiogenic agents, and may be applicable for cancer
    Platelet-derived growth
    factor subunit A
    (Isoform long)
    (SEQ ID NO: 257)
    Platelet-Derived Growth Vascular Endothelial Growth Factor VEGF activity can be determined using assays
    Factor B chain GeneSeq known in the art, such as those disclosed in International Publication No.
    Accession R38919 U.S. W00045835, for example. Promotion of growth and proliferation of cells, such as
    Pat. No. 5,219,739 vascular endothelial cells. Hematopoietic and immune dis- orders. Antagonists may
    P01127/PDGFB_HUMAN be useful as anti-angiogenic agents, and may be applicable for cancer
    Platelet-derived growth
    factor subunit B
    (isoform 1)
    (SEQ ID NO: 258)
    Stromal Derived Factor- 1 Stromal Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    alpha GeneSeq Accession Cancer Res. 48: 4266) Hematopoietic, immune disorders, cancer
    Y39995 WO9948528
    P48061-2/SDF1_HUMAN
    Isoform Alpha of Stromal
    cell-derived factor 1
    (isoform alpha)
    (SEQ ID NO: 259)
    Stromal Derived Factor- 1 Stromal Growth Factor Proliferation assay using NR6R-3T3 cells (Rizzino 1988
    beta GeneSeq Accession Cancer Res. 48: 4266) Hematopoietic, immune disorders, cancer
    R75420 CA2117953
    P48061/SDF1_HUMAN
    Stromal cell-derived factor 1
    (isoform beta)
    (SEQ ID NO: 260)
    Tarc GeneSeq Accession Chemotactic for T lymphocytes. May play a role in T-cell development. Thought to
    W14917 WO9711969 bind CCR8 and CCR4 Chemotactic leukocyte migration assays are known in the art,
    Q92583/CCL17_HUMAN for example: J. Immunol. Methods 33 ((1980)) Antiinflammatory. Immune disorders,
    C-C motif chemokine 17 cancer
    (SEQ ID NO: 261)
    Prolactin GeneSeq Prolactin is involved in immune cell proliferation and apoptosis. Immune coil
    Accession R78691 proliferation and suppression of apoptosis by prolactin can be assayed by methods
    WO9521625 well-known in the art, for example, Buckley, A R and Buckley D J, Ann NY Acad Sci
    P01236/PRL_HUMAN 2000; 917: 522-33, and within. Reproductive system disorders, cancer.
    Prolactin
    (SEQ ID NO: 262)
    Prolactin2 GeneSeq Prolactin is involved in immune cell proliferation and apoptosis. Immune coil
    Accession Y31764 U.S. proliferation and suppression of apoptosis by prolactin can be assayed by methods
    Pat. No. 5,955,346 well-known in the art, for example, Buckley, A R and Buckley D J, Ann NY Acad Sci
    2000; 917: 522-33, and within. Reproductive system disorders, cancer.
    Follicle stimulating FSH stimulates secretion of interleukin-1 by cells isolated from women in the follicular
    hormone Alpha subunit phase FSH activities can be determined using assays known in the art; J Gend Specif
    GeneSeq Accession Med 1999 November-December; 2(6): 30-4; Mol Cell Endocrinol. 1997 Nov. 15; 134(2): 109-18.
    Y54160 EP974359 Reproductive system disorders, cancer.
    P01215/GLHA_HUMAN
    Glycoprotein hormones
    alpha chain
    (SEQ ID NO: 263)
    Follicle stimulating FSH stimulates secretion of interleukin-1 by cells isolated from women in the follicular
    hormone Beta subunit phase FSH activities can be determined using assays known in the art; J Gend Specif
    GeneSeq Accession Med 1999 November-December; 2(6): 30-4; Mol Cell Endocrinol. 1997 Nov. 15; 134(2): 109-18.
    Y54161 EP974359 Reproductive system disorders, cancer.
    P01225/FSHB_HUMAN
    Follitropin subunit beta
    (SEQ ID NO: 264)
    Substance P (tachykinin) Substance P is associated with immunoregulation. Immuneregulation and bone
    GeneSeq Accession marrow, cell proliferation by substance P can be assayed by methods well-known in
    B23027 WO0054053 the art, for example, Lai et al. Proc Natl Acad Sci USA 2001 Mar. 27; 98(7): 3970-5;
    (SEQ ID NO: 385) Jallat-Daloz et al. Allergy Asthma Proc 2001 January-February; 22(1): 17-23; Kehler et al. Exp
    Lung Res 2001 January-February; 27(1): 25-46; and Adamus M A and Dabrowski Z J. J Cell
    Biochem 2001; 81(3)499-506. diabetes mellitus, hypertension, cancer
    Oxytocin (Neurophysin I) Oxytocin is involved in the induction of prostaglandin (E2) release as well as an
    GeneSeq Accession increased amount of calcium release by smooth muscle cells. Oxytocin and
    B24085 and B24086 prostaglandin E(2) release and Ocytocin (Ca2+) increase can be assayed by
    WO0053755 methods well-known in the art, for example, Pavan et al., AM J Obset Gynecol 2000
    P01178/NEU1_HUMAN July; 183(1): 76-82 and Holda et al., Cell Calcium 1996 July; 20(1): 43 51. inflammatory
    Oxytocin-neurophysin 1 disorders immunologic disorders, cancer
    (SEQ ID NO: 265)
    Vasopressin (Neurophysin Vasopressinis believed to have a direct antidiuretic action on the kidney, and it is
    II) GeneSeq Accession thought to cause vasoconstriction of the peripheral vessels. Vasopressin activity can
    B24085 and B24086 be determined using assays known in the art, for example, Endocr Regul 1996 March;
    WO0053755 30(I): 13-17. inflammatory disorders immunologic disorders, cancer
    P01185/NEU2_HUMAN
    Vasopressin-neurophysin
    2-copeptin
    (SEQ ID NO: 266)
    IL-1 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    P60326 EP165654 monocytes, and macrophages. Known functions include stimulating proliferation of
    IL-1 alpha immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P01583|IL1A_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-1 alpha can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 269) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    IL-1 beta D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
    P01584|IL1B_HUMAN inflammatory disorders immunologic disorders, cancer
    Interleukin-1 beta
    (SEQ ID NO: 267)
    IL-1 mature GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R14855 monocytes, and macrophages. Known functions include stimulating proliferation of
    EP456332 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    (mature truncated form of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    wherein the precursor is can be determined using assays known in the art: Matthews et al., in Lymphokines
    cleaved between amino and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    acids 116-117) D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
    (SEQ ID NO: 386) inflammatory disorders immunologic disorders, cancer
    IL-1 beta GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession Y08322 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9922763 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P01584|IL1B_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-1 beta can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 267) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    IL-3 variants GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession P80382, monocytes, and macrophages. Known functions include stimulating proliferation of
    P80383, P80384, and immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P80381 WO8806161 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Variants of wildtype IL-3 can be determined using assays known in the art: Matthews et al., in Lymphokines
    which has the sequence: and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    P08700|IL3_HUMAN D.C. 1987, pp. 221-225; and Kitamura et al (1989) J Cell Physiol. 140 323-334.
    Interleukin-3 inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 449)
    IL-4 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    P70615 WO8702990 monocytes, and macrophages. Known functions include stimulating proliferation of
    P05112/IL4_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Interleukin-4 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (isoform 1) can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 268) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    287-295. inflammatory disorders immunologic disorders, cancer
    IL-4 muteins GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession W52151 monocytes, and macrophages. Known functions include stimulating proliferation of
    W52152 W52153 W52154 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W52155 W52156 W52157 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    W52158 W52159 W52160 can be determined using assays known in the art: Matthews et al., in Lymphokines
    W52161 W52162 W52163 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    W52164 and W52165 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    WO9747744 287-295. inflammatory disorders immunologic disorders, cancer
    Variants of wildtype IL-4
    which has the sequence:
    P05112/IL4_HUMAN
    Interleukin-4
    (isoform 1)
    (SEQ ID NO: 268)
    IL-1 alpha GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession P90108 monocytes, and macrophages. Known functions include stimulating proliferation of
    EP324447 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P01583|IL1A_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-1 alpha can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 269) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    IL-3 variants GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R38561, monocytes, and macrophages. Known functions include stimulating proliferation of
    R38562, R38563, R38564, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    R38565, R38566, R38567, of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    R38568, R38569, R38570, can be determined using assays known in the art: Matthews et al., in Lymphokines
    R38571, and R38572 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    WO9307171 D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    Variants of wildtype IL-3 inflammatory disorders immunologic disorders, cancer
    which has the sequence:
    P08700|IL3_HUMAN
    Interleukin-3
    (SEQ ID NO: 449)
    IL-6 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    R45717 and R45718 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9402512 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P05231/IL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-6 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 270) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    inflammatory disorders immunologic disorders, cancer. Obesity. Metabolic Disease.
    Diabetes.
    IL-13 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    R48624 WO9404680 monocytes, and macrophages. Known functions include stimulating proliferation of
    P35225/IL13_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Interleukin-13 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (SEQ ID NO: 271) can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Boutelier et al (1995) J. Immunol. Methods 181, 29.
    inflammatory disorders immunologic disorders, cancer
    IL-4 mutein GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R47182 monocytes, and macrophages. Known functions include stimulating proliferation of
    DE4137333 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Variants of wildtype IL-4 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    which has the sequence: can be determined using assays known in the art: Matthews et al., in Lymphokines
    P05112/IL4_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    (isoform 1) 287-295. inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 268)
    IL-4 mutein Y124X Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R47183 DE4137333 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Variants of wildtype IL-4 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    which has the sequence: can be determined using assays known in the art: Matthews et al., in Lymphokines
    P05112/IL4_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    (isoform 1) 287-295. inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 268))
    IL-4 mutein Y124G Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R47184 DE4137333 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Variants of wildtype IL-4 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    which has the sequence: can be determined using assays known in the art: Matthews et al., in Lymphokines
    P05112/IL4_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    (isoform 1) 287-295. inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 268)
    Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    (precursor) GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R41664 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9317698 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P22301/IL10_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-10 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (precursor form is D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
    processed into a truncated inflammatory disorders immunologic disorders, cancer
    mature form)
    (SEQ ID NO: 272)
    Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R42642 WO9318783-A immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 3 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9318783-A can be determined using assays known in the art: Matthews et al., in Lymphokines
    (mature IL-10) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 273) D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
    inflammatory disorders immunologic disorders, cancer
    Human interleukin-1 beta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    precursor. GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R42447 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP569042 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P01584/IL1B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-1 beta and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 274) D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    Interleukin- 1alpha Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R45364 EP578278 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P01583|IL1A_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-1 alpha can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 269) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    Human interleukin-3 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    variant GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R22814 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    JP04063595 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Variants of wildtype IL-3 can be determined using assays known in the art: Matthews et al., in Lymphokines
    which has the sequence: and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    P08700|IL3_HUMAN D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
    Interleukin-3 inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 449)
    IL-1i fragments GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R35484 and monocytes, and macrophages. Known functions include stimulating proliferation of
    R35485 EP541920 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    IL-1 inhibitor (IL-li) Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R35486 and R35484 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EPS541920 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarclio (1989) inflammatory disorders
    immunologic disorders, cancer
    ICE
    22 kD subunit. Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R33780 EP533350 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 16 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    EP533350 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 450) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    ICE
    20 kD subunit. Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R33781 EP533350 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 17 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    EP533350 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 451) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    ICE
    10 kD subunit Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R33782 EP533350 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 18 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    EP533350 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 452) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    (precursor) GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R41664 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9317698 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P22301/IL10_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-10 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (precursor form is D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
    processed into a truncated inflammatory disorders immunologic disorders, cancer
    mature form)
    (SEQ ID NO: 272)
    Human Interleukin-10 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R42642 WO9318783 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 3 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9318783-A can be determined using assays known in the art: Matthews et al., in Lymphokines
    (mature IL-10) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 273) D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
    inflammatory disorders immunologic disorders, cancer
    Human Interleukin-1 beta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    precursor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R42447 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP569042 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P01584/IL1B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-1 beta and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 274) D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
    inflammatory disorders immunologic disorders, cancer
    Human interleukin-6 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R49041 WO9403492 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P05231/IL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-6 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 270) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    inflammatory disorders immunologic disorders, cancer
    Mutant Interleukin 6 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    S176R GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R54990 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9411402 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    S176R variant of wildtype can be determined using assays known in the art: Matthews et al., in Lymphokines
    IL-6 which has the and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    sequence: D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    P05231/IL6_HUMAN inflammatory disorders immunologic disorders, cancer
    Interleukin-6
    (SEQ ID NO: 270)
    Interleukin 6 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R55256 monocytes, and macrophages. Known functions include stimulating proliferation of
    JP06145063 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P05231/IL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-6 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 270) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    inflammatory disorders immunologic disorders, cancer
    Interleukin 8 (IL-8) Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R53932 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    JP06100595 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    GenBank: AAA59159.1 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 275) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8
    receptor polypep- tides may be useful for inhibiting interleukin activities.
    Human interleukin-7 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R59919 U.S. Pat. No. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    5,328,988 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P13232/IL7_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-7 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; and Park et al (1990) J. Exp. Med. 171, 1073-79.
    (SEQ ID NO: 276) inflammatory disorders immunologic disorders, cancer
    IL-3 containing fusion Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein. GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R79342 and immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    R79344 WO9521254 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Fusions of wildtype IL-3 can be determined using assays known in the art: Matthews et al., in Lymphokines
    which has the sequence: and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    P08700|IL3_HUMAN D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
    Interleukin-3 inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 449)
    IL-3 mutant proteins Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R79254, R79255, R79256, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    R79257, R79258, R79259, of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    R79260, R79261, R79262, can be determined using assays known in the art: Matthews et al., in Lymphokines
    R79263, R79264, R79265, and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    R79266, R79267, R79268, D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830. inflammatory
    R79269, R79270, R79271, disorders immunologic disorders, cancer
    R79272, R79273, R79274,
    R79275, R79276, R79277,
    R79278, R79279, R79280,
    R79281, R79282, R79283,
    R79284, and R79285
    ZA9402636
    Variants of wildtype IL-3
    which has the sequence:
    P08700|IL3_HUMAN
    Interleukin-3
    (SEQ ID NO: 449)
    IL-12 p40 subunit. Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R63018 AU9466072 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P2946/|IL12B_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-12 subunit beta can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 277) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    AGF GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    R64240 WO9429344 monocytes, and macrophages. Known functions include stimulating proliferation of
    Q8NI99/ANGL6_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Angiopoietin-related of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    protein
    6 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 278) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    Human interlaukin-12 40 kD Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    subunit GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R79187 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9519786 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P2946/|IL12B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-12 subunit beta and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 277) D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. inflammatory
    disorders immunologic disorders, cancer
    Human interleukin-15 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor from clone P1 monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    R90843 WO9530695 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q13261|I15RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-15 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit alpha D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830. Soluble IL-8
    Isoform 1) receptor polypep- tides may be useful for inhibiting interleukin activities. Obesity.
    (SEQ ID NO: 453) Metabolic Disease. Diabetes. Enhancing secretion and stability of Interleukin 15
    Human interleukin-7 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R92796 WO9604306 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P13232/IL7_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-7 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (isoform 1) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 276) D.C. 1987, pp. 221-225; and Park et al (1990) J. Exp. Med. 171, 1073-79.
    inflammatory disorders immunologic disorders, cancer
    interleukin-9 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R92797 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9604306 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P15248/IL9_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-9 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 279) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. inflammatory
    disorders immunologic disorders, cancer
    interleukin-3 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R92801 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9604306 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P08700|IL3_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-3 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 280) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
    inflammatory disorders immunologic disorders, cancer
    Human interleukin-5 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R92802 WO9604306 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P05113/IL5_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-5 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 281) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334.
    inflammatory disorders immunologic disorders, cancer
    Recombinant interleukin- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    16 GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    W33373 DE19617202 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q14005/IL16_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Pro-interleukin-16 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (isoform 1) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 282) D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70. inflammatory
    disorders immunologic disorders, cancer
    Human IL-16 protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    W33234 DE19617202 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q14005/IL16_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Pro-interleukin-16 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (isoform 1) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 282) D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70. inflammatory
    disorders immunologic disorders, cancer
    Thrl 17 human interleukin Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    9 GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    W27521 WO9708321 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P15248|IL9_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-9 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 387) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    Metl 17 human interleukin Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    9 GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    W27522 WO9708321 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    (SEQ ID NO: 388) of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. inflammatory
    disorders immunologic disorders, cancer
    Human intracellular IL-1 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor antagonist. monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W77158 EP864585 (e.g. of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NOs: 12 to 19, or can be determined using assays known in the art: Matthews et al., in Lymphokines
    22 to 25 of this and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    publication). D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    Human interleukin-18 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein (IL-18) GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W77158 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP864585 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q14116/IL18_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-18 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; and USHIO et al (1996) J. Immunol. 156, 4274-79.
    (SEQ ID NO: 283) inflammatory disorders immunologic disorders, cancer
    Human interleukin-18 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    W77077 EP861663 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q14116/IL18_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-18 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (isoform 1) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 283) D.C. 1987, pp. 221-225; and USHIO et al (1996) J. Immunol. 156, 4274-79.
    inflammatory disorders immunologic disorders, cancer
    Human interleukin 18 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    derivatives GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accessions W77083, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W77084, W77085, of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    W77086, W77087, can be determined using assays known in the art: Matthews et al., in Lymphokines
    W77088, and W77089 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    EP861663 D.C. 1987, pp. 221-225; and Ushio et al (1996) J. Immunol, 156, 4274-79.
    Variants of wildtype IL18 inflammatory disorders immunologic disorders, cancer
    which is provided as:
    Q14116/IL18_HUMAN
    Interleukin-18
    (isoform 1)
    (SEQ ID NO: 283)
    Interleukin-9 (IL-9) mature Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein (Thr117 version). monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W68158 WO9827997 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    FIG. 2 of WO9827997 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 389) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. inflammatory
    disorders immunologic disorders, cancer
    IL-9 mature protein variant Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    (Met117 version) GenSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W68157 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9827997 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    FIG. 3 of WO9827997 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 390) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. inflammatory
    disorders immunologic disorders, cancer
    Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein variant #3. monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W64058 WO9824904 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Wildtype IL-9R is provided can be determined using assays known in the art: Matthews et al., in Lymphokines
    as: and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Q01113/IL9R_HUMAN D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. inflammatory
    Interleukin-9 receptor disorders immunologic disorders, cancer
    (isoform 1)
    (SEQ ID NO: 303)
    Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein variant fragment monocytes, and macrophages. Known functions include stimulating proliferation of
    GenSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W64060 WO9824904 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Wildtype IL-9R is provided can be determined using assays known in the art: Matthews et al., in Lymphokines
    as: and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Q01113/IL9R_HUMAN D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. Soluble IL-9
    Interleukin-9 receptor receptor polypep- tides may be useful for inhibiting interleukin activities.
    (isoform 1)
    (SEQ ID NO: 303)
    Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein variant #3. monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W64061 WO9824904 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Wildtype IL-9R is provided can be determined using assays known in the art: Matthews et al., in Lymphokines
    as: and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Q01113/IL9R_HUMAN D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. Soluble IL-9
    Interleukin-9 receptor receptor polypep- tides may be useful for inhibiting interleukin activities.
    (isoform 1)
    (SEQ ID NO: 303)
    Human Interleukin-12 p40 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W51311 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9817689 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P2946/|IL12B_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-12 subunit beta and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 277) D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. inflammatory
    disorders immunologic disorders, cancer
    Human Interleukin-12 p35 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W51312 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9817689 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P29459/IL12A_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-12 subunit and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    alpha D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. inflammatory
    (SEQ ID NO: 284) disorders immunologic disorders, cancer
    Human protein with IL-16 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    activity GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W63753 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    DE19649233- of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70. inflammatory
    disorders immunologic disorders, cancer
    Human protein with IL-16 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    activity GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W59425 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    DE19649233- of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Lim et al (1996) J. Immunol. 156, 2566-70. inflammatory
    disorders immunologic disorders, cancer
    Human interleukin-15 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    W53878 U.S. Pat. No. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    5,747,024 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P40933/IL15_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-15 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform IL15-S48AA) D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830. inflammatory
    (SEQ ID NO: 285) disorders immunologic disorders, cancer. Obesity. Metabolic Disease. Diabetes.
    Human wild-type Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    interleukin-4 (hIL-4) monocytes, and macrophages. Known functions include stimulating proliferation of
    protein GeneSeq immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Accession W52149 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9747744 can be determined using assays known in the art: Matthews et al., in Lymphokines
    P05112/IL4_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-4 D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    (isoform 1) 287-295. inflammatory disorders immunologic disorders, cancer
    (SEQ ID NO: 286)
    interleukin-4 muteins Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accessions monocytes, and macrophages. Known functions include stimulating proliferation of
    W52150, W52151, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W52153, W52154, of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    W52155, W52156, can be determined using assays known in the art: Matthews et al., in Lymphokines
    W52157, W52158, and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    W52159, W52160, D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    W52161, W52162, 287-295. inflammatory disorders immunologic disorders, cancer
    W52163, W52164,
    W52165, W52166, and
    W52167 WO9747744
    Variants of wildtype IL-4
    which has the sequence:
    P05112/IL4_HUMAN
    Interleukin-4
    (isoform 1)
    (SEQ ID NO: 268)
    Human interleukin 1 delta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    Y28408 WO9935268 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 4 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9935268 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 287) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    Human interleukin-1 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor antagonist beta monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Y24395 WO9935268 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
    inflammatory disorders immunologic disorders, cancer
    Human EDIRF II protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    sequence GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession Y22199 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9932632 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 6 of can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9932632 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 391) D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    Human EDIRF I protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    sequence GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession Y22197 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9932632 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 2 of can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9932632 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 392) D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    Human IL- 1RD10 protein Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    sequence GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession Y14131 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9919480 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 20 of can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9919480 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20. Soluble
    IL-1RD10 receptor polypep- tides may be useful for inhibiting interleukin activites.
    Human IL- 1RD9 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    Y14122 WO9919480 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NOS: 6, 8, 10 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9919480 can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20. Soluble
    IL-1RD10 receptor polypep- tides may be useful for inhibiting interleukin activites.
    Human DNAX interleukin- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    40 GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    Y09196 WO9919491 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    SEQ ID NO: 2 or 4 of of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9919491 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 454) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    (DIL-40) alternative Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    sequence GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession Y09197 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9919491 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 4 of can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9919491 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 455) D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    IL-11 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    R50176 WO9405318 monocytes, and macrophages. Known functions include stimulating proliferation of
    P2080/|IL11_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Interleukin-11 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (isoform 1) can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 288) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Lu et al (1994) J immunol. Methods 173, 19.
    inflammatory disorders immunologic disorders, cancer
    Human adipogenesis Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    inhibitory factor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R43260 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP566410 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (also known as IL-11) can be determined using assays known in the art: Matthews et al., in Lymphokines
    P2080/|IL11_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-11 D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    (isoform 1)
    (SEQ ID NO: 288)
    IL-11 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    W02202 JP08127539 monocytes, and macrophages. Known functions include stimulating proliferation of
    P2080/|IL11_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Interleukin-11 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (isoform 1) can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 288) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Lu et al (1994) J immunol. Methods 173, 19.
    inflammatory disorders immunologic disorders, cancer
    IL-14 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    R55800 WO9416074 monocytes, and macrophages. Known functions include stimulating proliferation of
    P40222/TXLNA_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Alpha-taxilin of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (SEQ ID NO: 289) can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Ambrus et al (1993) PNAS 90, 63330-34. inflammatory
    disorders immunologic disorders, cancer
    IL-17 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession B03807 U.S. monocytes, and macrophages. Known functions include stimulating proliferation of
    Pat. No. 6,072,033 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q96F46/I17RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-17 receptor A can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 290) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86. Soluble IL-
    17 receptor polypep- tides may be useful for inhibiting interleukin activites.
    IL-17 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    R76573 WO9518826 monocytes, and macrophages. Known functions include stimulating proliferation of
    Q16552/IL17_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Interleukin-17A of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (SEQ ID NO: 291) can be determined using assays known in the art: Matthews et al., in Lymphokines
    and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86. inflammatory
    disorders immunologic disorders, cancer
    CTLA-8 GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession W13651 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9704097 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    (also known as IL-17) of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q16552/IL17_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-17A and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 291) D.C. 1987, pp. 221-225. inflammatory disorders immunologic disorders, cancer
    IL-19 GeneSeq Accession Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    W37935 WO9808870 monocytes, and macrophages. Known functions include stimulating proliferation of
    Q9UHD0|IL19_HUMAN immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Interleukin-19 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    (isoform 1) can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 292) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Gallagher et al (2000) Genes Immun. 1,442-50.
    inflammatory disorders immunologic disorders, cancer
    IL-21 (TIF) GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession Y92879 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO0024758 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q9HBE4/IL21_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-21 can be determined using assays known in the art: Matthews et al., in Lymphokines
    (isoform 1) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 293) D.C. 1987, pp. 221-225; and Parrish-Novak et al (2000) Nature 408, 57-63.
    inflammatory disorders immunologic disorders, cancer
    IL-8 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R33420 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9306229 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    IL-8RA of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P25024/CXCR1_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    C-X-C chemokine receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    type 1 D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8
    (SEQ ID NO: 294) receptor polypep- tides may be useful for inhibiting interleukin activities.
    IL-8RB
    P25025/CXCR2_HUMAN
    C-X-C chemokine receptor
    type 2
    (SEQ ID NO: 295)
    Human type II interleukin- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    1 receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R85480 U.S. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Pat. No. 5,464,937 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P27930/IL1R2_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-1 receptor type 2 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 296) D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20. Soluble
    type II interleukin-1 receptor polypep- tides may be useful for inhibiting interleukin
    activities.
    Human interleukin-12 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R69632 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP638644 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    IL-12 receptor B1 can be determined using assays known in the art: Matthews et al., in Lymphokines
    P42701|I12R1_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-12 receptor D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. Soluble IL-12
    subunit beta-1 receptor polypep- tides may be useful for inhibiting interleukin activities.
    (isoform 1)
    (SEQ ID NO: 393)
    IL-12 receptor B2
    Q99665|I12R2_HUMAN
    Interleukin-12 receptor
    subunit beta-2
    (isoform 1)
    (SEQ ID NO: 394)
    Interleukin 8 receptor B Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R80758 U.S. Pat. No. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    5,440,021 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    IL-8RB can be determined using assays known in the art: Matthews et al., in Lymphokines
    P25025/CXCR2_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    C-X-C chemokine receptor D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8
    type 2 receptor B polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 295)
    Human IL-8 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein hIL8RA GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession B09989 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    JP08103276 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    IL-8RA can be determined using assays known in the art: Matthews et al., in Lymphokines
    P25024/CXCR1_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    C-X-C chemokine receptor D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8
    type 1 receptor A polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 294)
    Human IL-8 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein hIL8R GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession B09990 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    JP08103276 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    IL-8RA can be determined using assays known in the art: Matthews et al., in Lymphokines
    P25024/CXCR1_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    C-X-C chemokine receptor D.C. 1987, pp. 221-225; and Holmes et al (1991) Science 253, 1278-80. Soluble IL-8
    type 1 receptor polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 294)
    IL-8RB
    P25025/CXCR2_HUMAN
    C-X-C chemokine receptor
    type 2
    (SEQ ID NO: 295)
    Interleukin-2 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    associated protein p43 monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    R97569 WO9621732- of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 2 of can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9621732 and Interferons: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 395) D.C. 1987, pp. 221-225; and Gillis et al (1978) J. Immunol. 120, 2027. Soluble IL-2
    receptor polypep- tides may be useful for inhibiting interleukin activities.
    Human interleukin-17 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W04185 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9629408 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q96F46/I17RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-17 receptor A and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 290) D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86. Soluble IL-
    17 receptor polypep- tides may be useful for inhibiting interleukin activities.
    Human interleukin-11 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R99090 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9619574 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q14626/I11RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-11 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit alpha D.C. 1987, pp. 221-225; and Lu et al (1994) J immunol. Methods 173, 19. Soluble IL-
    (SEQ ID NO: 297) 11 receptor polypep- tides may be useful for inhibiting interleukin activities.
    Human interleukin-1 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor accessory protein monocytes, and macrophages. Known functions include stimulating proliferation of
    GeneSeq Accession immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W01911 WO9623067 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Human IL1R Acp can be determined using assays known in the art: Matthews et al., in Lymphokines
    SEQ ID NO: 3 of and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    WO9623067 D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20.
    (SEQ ID NO: 396) Inflammatory disorders immunologic disorders, cancer
    Soluble Human IL1R Acp
    SEQ ID NO: 9 of
    WO9623067
    (SEQ ID NO: 397)
    AGF Protein GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R92749 U.S. monocytes, and macrophages. Known functions include stimulating proliferation of
    Pat. No. 5,488,032 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q8NI99/ANGL6_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Angiopoietin-related can be determined using assays known in the art: Matthews et al., in Lymphokines
    protein
    6 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 278) D.C. 1987, pp. 221-225. Inflammatory disorders immunologic disorders, cancer
    Human interleukin-1 type- Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    3 receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R91064 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W09607739 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 2 and 4 of can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9607739 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 398 and SEQ D.C. 1987, pp. 221-225; and Orencole & Dinarello (1989) Cytokine 1, 14-20. Soluble
    ID NO: 399, respectively) IL-type-3 receptor polypep- tides may be useful for inhibiting interleukin activities
    Human interleukin-13 beta Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W24972 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9720926 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    SEQ ID NO: 2 from can be determined using assays known in the art: Matthews et al., in Lymphokines
    WO9720926 and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (SEQ ID NO: 400) D.C. 1987, pp. 221-225; and Boutelier et al (1995) J. Immunol. Methods 181, 29.
    Soluble IL-13 beta receptor polypep- tides may be useful for inhibiting interleukin
    activities.
    Human interleukin-13 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    alpha receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W24973 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    WO9720926 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    IL-13RA1 can be determined using assays known in the art: Matthews et al., in Lymphokines
    P78552/I13R1_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-13 receptor D.C. 1987, pp. 221-225; and Boutelier et al (1995) J. Immunol. Methods 181, 29.
    subunit alpha-1 Soluble IL-13 alpha receptor polypep- tides may be useful for inhibiting interleukin
    (isoform 1) activities.
    (SEQ ID NO: 298)
    IL-13RA2
    Q14627/I13R2_HUMAN
    Interleukin-13 receptor
    subunit alpha-2
    (SEQ ID NO: 299)
    Human interleukin-4 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W13499 U.S. immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Pat. No. 5,599,905 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P24394/IL4RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-4 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit alpha D.C. 1987, pp. 221-225; and Siegel & Mostowski (1990) J Immunol Methods 132,
    (isoform 1) 287-295. Soluble IL-4 receptor polypep- tides may be useful for inhibiting interleukin
    (SEQ ID NO: 300) activities.
    Human interleukin-12 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    beta-2 receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W12771 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP759466 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q9966/|I12R2_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-12 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit beta-2 D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. Soluble IL-12
    (isoform 1) beta-2 receptor polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 301)
    Human interleukin-12 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    beta-1 receptor. GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession W12772 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP759466 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P4270/|I12R1_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-12 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit beta-1 D.C. 1987, pp. 221-225; and Hori et al (1987), Blood 70, 1069-1078. Soluble IL-12
    (isoform 1) beta-1 receptor polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 302)
    Human IL-9 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    protein GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accessions W64055, immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    W64056, and W64057 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    WO9824904 can be determined using assays known in the art: Matthews et al., in Lymphokines
    Q01113/IL9R_HUMAN and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    Interleukin-9 receptor D.C. 1987, pp. 221-225; and Yang et al (1989) Blood 74, 1880-84. Soluble IL-9
    (isoform 1) receptor polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 303)
    IL-10 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession W41804 U.S. monocytes, and macrophages. Known functions include stimulating proliferation of
    Pat. No. 5,716,804 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    IL-10RA of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Q13651/I10R1_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-10 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit alpha D.C. 1987, pp. 221-225; and Thompson-Snipes et al (1991) J. Exp. Med. 173, 507-510.
    (SEQ ID NO: 304) Soluble IL-10 receptor polypep- tides may be useful for inhibiting interleukin
    IL-10RB activities.
    Q0833/|I10R2_HUMAN
    Interleukin-10 receptor
    subunit beta
    (SEQ ID NO: 305)
    Human IL-6 receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    Y30938 JP11196867 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P08887/IL6RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-6 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines
    subunit alpha and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    (SEQ ID NO: 306) Soluble IL-6 receptor polypep- tides may be useful for inhibiting interleukin activities.
    II-17 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession Y97181 U.S. monocytes, and macrophages. Known functions include stimulating proliferation of
    Pat. No. 6,096,305 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q96F46/I17RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-17 receptor A can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 290) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86. Soluble IL-
    17 receptor polypep- tides may be useful for inhibiting interleukin activities.
    II-17 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession Y97131 U.S. monocytes, and macrophages. Known functions include stimulating proliferation of
    Pat. No. 6,100,235 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q96F46/I17RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-17 receptor A can be determined using assays known in the art: Matthews et al., in Lymphokines
    (SEQ ID NO: 290) and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    D.C. 1987, pp. 221-225; and Yao et al (1995) J. Immunol. 155, 5483-86. Soluble IL-
    17 receptor polypep- tides may be useful for inhibiting interleukin activities.
    Human interleukin-3 Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession R25300 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    EP509826 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P26951/IL3RA_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-3 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit alpha D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334. Soluble
    (isoform 1) IL-3 receptor polypep- tides may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 307)
    Human GM- CSF receptor Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R10919 WO9102063 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    GM-CSF receptor A of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P15509/CSF2R_HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Granulocyte-macrophage and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    colony-stimulating factor D.C. 1987, pp. 221-225. Soluble GM-CSF receptor polypep- tides may be useful for
    receptor subunit alpha inhibiting interleukin activities.
    (isoform 1)
    (SEQ ID NO: 308)
    GM-CSF receptor B
    P32927/IL3RB_HUMAN
    Cytokine receptor
    common subunit beta
    (isoform 1)
    (SEQ ID NO: 309)
    Human IL-5 receptor alpha Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    chain GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R25064 EP492214 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q01344/IL5RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-5 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines
    subunit alpha and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334. Soluble
    (SEQ ID NO: 310) IL-5 receptor alpha polypeptides may be useful for inhibiting interleukin activities.
    II-5 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession W82842 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9847923 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q01344/IL5RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-5 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines
    subunit alpha and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; Kitamura et al (1989) J Cell Physiol. 140 323-334. Soluble
    (SEQ ID NO: 310) IL-5 receptor polypep- tides may be useful for inhibiting interleukin activities.
    II-6 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R37215 monocytes, and macrophages. Known functions include stimulating proliferation of
    JP05091892 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P08887/IL6RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-6 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines
    subunit alpha and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; and Aarden et al (1987) Eur. J. Immunol 17, 1411-16.
    (SEQ ID NO: 306) Soluble IL-6 receptor polypep- tides may be useful for inhibiting interleukin activities.
    Human B cell stimulating Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    factor-2 receptor GeneSeq monocytes, and macrophages. Known functions include stimulating proliferation of
    Accession P90525 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    AU8928720 of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    P08887/IL6RA HUMAN can be determined using assays known in the art: Matthews et al., in Lymphokines
    Interleukin-6 receptor and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    subunit alpha D.C. 1987, pp. 221-225. Soluble B cell stimulating factor-2 receptor polypep- tides
    (isoform 1) may be useful for inhibiting interleukin activities.
    (SEQ ID NO: 306)
    IL-7 receptor clone Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    GeneSeq Accession monocytes, and macrophages. Known functions include stimulating proliferation of
    R08330 EP403114 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    P1687/|IL7RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-7 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines
    subunit alpha and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; and Park et al (1990) J. Exp. Med. 171, 1073-79. Soluble IL-
    (SEQ ID NO: 311) 7 receptor polypep- tides may be useful for inhibiting interleukin activities.
    EPO receptor; EPOR EPO Receptor is involved in the proliferation and differentiation of erythroblasts. EPO
    GeneSeq Accession Receptor activity can be determined using assays known in the art, such as, J Biol
    R06512 WO9008822 Chem 2001 Mar. 23; 276(12: 8995-9002; JAK2 protein tyrosine kinase activity: Blood
    P19235/EPOR_HUMAN 1994 Sep. 1; 84(5): 1501-7 and Mol Cell Biol. 1994 October; 14(10: 6506-14. Inflammatory
    Erythropoietin receptor disorders, immunologic disorders, cancer, erythroblast proliferation and differentiation
    (isoform EPOR-F)
    (SEQ ID NO: 312)
    IL-15 receptor GeneSeq Interleukins are a group of multi- functional cytokines synthesized by lymphocytes,
    Accession R90843 monocytes, and macrophages. Known functions include stimulating proliferation of
    WO9530695 immune cells (e.g., T helper cells, B cells, eosinophils, and lymphocytes), chemotaxis
    Q1326/|I15RA_HUMAN of neutrophils and T lymphocytes, and/or inhibition of interferons. Interleukin activity
    Interleukin-15 receptor can be determined using assays known in the art: Matthews et al., in Lymphokines
    subunit alpha and Interferens: A Practical Approach, Clemens et al., eds, IRL Press, Washington,
    (isoform 1) D.C. 1987, pp. 221-225; and Giri et al (1994) EMBO J. 13 2822-2830. Soluble IL-15
    (SEQ ID NO: 313) receptor polypep- tides may be useful for inhibiting interleukin activities. Obesity.
    Metabolic Disease. Diabetes. Enhancing secretion and stability of Interleukin 15
    CD137; 4-1BB Receptor Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Protein GeneSeq cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    Accession R70977 stimulation can be determined using assays known in the art: Moore et al., 1999,
    WO9507984 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Q07011/TNR9_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble 4-1BB
    Tumor necrosis factor receptor polypeptides may be useful for inhibiting apoptosis, NF-kB activation,
    receptor superfamily and/or co-stimulation of immune cells such as B and T cells.
    member 9
    (SEQ ID NO: 314)
    BCMA GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession Y71979 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO0068378 stimulation can be determined using assays known in the art: Moore et al., 1999,
    Q02223/TNR17_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble BCMA
    receptor superfamily receptor polypeptides may be useful for inhibiting apoptosis, NF-kB activation,
    member 17 and/or co-stimulation of immune cells such as B and T cells.
    (isoform 1)
    (SEQ ID NO: 315)
    CD27 GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    R20814 WO9201049 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    P26842/CD27_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    CD27 antigen Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    (SEQ ID NO: 316) 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble CD27
    polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co-
    stimulation of immune cells such as B and T cells.
    CD30 GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    R35478 DE4200043 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    P28908/TNR8_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble CD30
    member
    8 polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co-
    (isoform 1) stimulation of immune cells such as B and T cells.
    (SEQ ID NO: 317)
    CD40 GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Y33499 WO9945944 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    P25942/TNR5_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble CD40
    member
    5 polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co-
    (isoform 1) stimulation of immune cells such as B and T cells.
    (SEQ ID NO: 318)
    EDAR Genbank Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession AAD50077 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    Q9UNE0|EDAR_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Immune Disorders,
    member EDAR Lymphomas, X-linked hypohidrotic ectodermal dysplasia
    (isoform 1)
    (SEQ ID NO: 319)
    OX40; ACT-4 GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R74737 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9512673 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P43489/TNR4_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Immune Disorders,
    receptor superfamily Lymphomas, T cell disorders
    member 4
    (SEQ ID NO: 320)
    TACI GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    W75783 WO9839361 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    O14836/TR13B_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    receptor superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble TACI
    member 13B receptor polypep- tides may be useful for inhibiting apoptosis, NF-kB activation,
    (isoform 1) and/or co-stimulation of immune cells such as B and T cells.
    (SEQ ID NO: 321)
    TNF-R GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R10986 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    AU9058976 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P19438/TNR1A_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble TNF-R
    receptor superfamily receptor polypep- tides may be useful for inhibiting apoptosis, NF-kB activation,
    member 1A and/or co-stimulation of immune cells such as B and T cells.
    (isoform 1)
    (SEQ ID NO: 322)
    TNF-RII; TNF p75 Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    receptor; Death Receptor cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    GeneSeq Accession stimulation can be determined using assays known in the art: Moore et al., 1999,
    R11141 EP418014 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    P20333/TNR1B_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble TNFR-II
    Tumor necrosis factor receptor polypep- tides may be useful for inhibiting apoptosis, NF-kB activation,
    receptor superfamily and/or co-stimulation of immune cells such as B and T cells.
    member 1B
    (isoform 1)
    (SEQ ID NO: 323)
    hAPO-4; TROY GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession W93581 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9911791 stimulation can be determined using assays known in the art: Moore et al., 1999,
    Q9N568/TNR19_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Immune Disorders,
    receptor superfamily Cancers
    member 19
    (isoform 1)
    (SEQ ID NO: 324)
    TNF-alpha precursor Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    GeneSeq Accession cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    P60074 EP205038 stimulation can be determined using assays known in the art: Moore et al., 1999,
    Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    disorders immunologic disorders, cancer
    Human TNF- alpha Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    GeneSeq Accession cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    R62463 EP619372 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P01375/TNFA_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    (SEQ ID NO: 325) disorders immunologic disorders, cancer
    Human TNF- alpha Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    GeneSeq Accession cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    R42679 EP563714 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P01375/TNFA_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    (SEQ ID NO: 325) disorders immunologic disorders, cancer
    Human TNF- beta (LT- Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    alpha) GeneSeq cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    Accession B37799 stimulation can be determined using assays known in the art: Moore et al., 1999,
    WO0064479 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    P01374/TNFB_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    Lymphotoxin-alpha disorders immunologic disorders, cancer
    (SEQ ID NO: 326)
    LT-alpha GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession P70107 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    EP250000 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P01374/TNFB_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Lymphotoxin-alpha 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    (SEQ ID NO: 326) disorders immunologic disorders, cancer
    LT-beta GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R56869 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9413808 stimulation can be determined using assays known in the art: Moore et al., 1999,
    Q06643/TNFC_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Lymphotoxin-beta 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    (isoform 1) disorders immunologic disorders, cancer
    (SEQ ID NO: 327)
    OPGL GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession W83195 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9846751 stimulation can be determined using assays known in the art: Moore et al., 1999,
    O14788/TNF11_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    ligand superfamily disorders immunologic disorders, cancer, loss of bone mass
    member 11
    (isoform 1)
    (SEQ ID NO: 328)
    FasL GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    W98071 WO9903999 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    P48023/TNFL6_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    ligand superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    member
    6 disorders immunologic disorders, cancer
    (isoform 1)
    (SEQ ID NO: 329)
    FasL GeneSeq Accession Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    W95041 WO9903998 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    P48023/TNFL6_HUMAN stimulation can be determined using assays known in the art: Moore et al., 1999,
    Tumor necrosis factor Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    ligand superfamily 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    member
    6 disorders immunologic disorders, cancer
    (isoform 1)
    (SEQ ID NO: 329)
    CD27L GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R50121 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9405691 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P32970/CD70_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    CD70 antigen 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    (isoform 1) disorders immunologic disorders, cancer
    (SEQ ID NO: 330)
    CD30 ligand GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R45007 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9324135 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P32971/TNFL8_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    ligand superfamily disorders immunologic disorders, cancer
    member 8
    (SEQ ID NO: 331)
    CD40L GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R85486 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9529935 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P29965/CD40L_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    CD40 ligand 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    (SEQ ID NO: 332) disorders immunologic disorders, cancer
    4-1BB ligand GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession W26657 U.S. cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    Pat. No. 5,674,704 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P41273/TNFL9_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    ligand superfamily disorders immunologic disorders, cancer
    member 9
    (SEQ ID NO: 333)
    FAS Ligand Inhibitory Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Protein (DcR3) GeneSeq cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    Accession B19335 stimulation can be determined using assays known in the art: Moore et al., 1999,
    WO0058465 Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    O95407/TNF6B_HUMAN 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Soluble DcR3
    Tumor necrosis factor polypeptides may be useful for inhibiting apoptosis, NF-kB activation, and/or co-
    receptor superfamily stimulation of immune cells such as B and T cells.
    member 6B
    (SEQ ID NO: 334)
    OX40L GeneSeq Activities associated with apoptosis, NF-kB activation, and co- stimulation of immune
    Accession R79903 cells such as T and B cells. Apoptosis activity, NF-kB activation, and B and T cell co-
    WO9521915 stimulation can be determined using assays known in the art: Moore et al., 1999,
    P23510/TNFL4_HUMAN Science, 285(5425): 260-3; Song H Y et al., 1997 Proc Natl Acad Sci USA 94(18):
    Tumor necrosis factor 9792-6; Epsevik and Nissen-Meyer, 1986, J. Immunol. Methods. Inflammatory
    ligand superfamily disorders immunologic disorders, cancer
    member 4
    (isoform 1)
    (SEQ ID NO: 335)
    Protease inhibitor peptides Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    GeneSeq Accessions the art: HIV protease assays: EP0387231. One can modify the assay to look for
    R12435, R12436, R12437, inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    R12438, R12439, R12440, inflammatory disorders, immuno- logic disorders, cancer, viral infections
    and R1244 WO9106561
    Retroviral protease Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    inhibitors GeneSeq the art: HIV protease assays: EP0387231. One can modify the assay to look for
    Accessions R06660, inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    R06661, R06662, R06663, inflammatory disorders, immuno- logic disorders, cancer, viral infections
    R06664, R06665, R06666,
    R06667, R06668, R06669,
    R06670, R06671, R06672,
    R06673, R06674, R06675,
    and R06676 EP387231
    HIV protease inhibiting Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    peptides GeneSeq the art: HIV protease assays: EP0387231. One can modify the assay to look for
    Accessions R59293, inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    R59294, R59295, R59296, inflammatory disorders, immuno- logic disorders, cancer, viral infections
    R59297, R59298, R59299,
    R592300, R59301,
    R59302, R59301, R59302,
    R59303, R59304, R59305,
    R59306, R59307, R59308,
    R59309, R59310, R59311,
    R59312, R59313, R59314,
    R59315, R59316, R59317
    R59318, R59319, R59320,
    R59321, R59322, R59323,
    R59324, R59325, R59326,
    R59327, R59328, R59329,
    R59330, R59331, R59332,
    R59333, R59334, R59335,
    R59336, R59337, R59338,
    R59339, R59340, R59341,
    R59342, R59343, R59344,
    R59345, R59346, R59347,
    R59348, R59349, and
    R59350 WO9301828
    HIV-1 protease inhibitors Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    GeneSeq Accessions the art: HIV protease assays: EP0387231. One can modify the assay to look for
    R86326, R86327, R86328, inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    R86329, R86330, R86331, inflammatory disorders, immuno- logic disorders, cancer, viral infections
    R86332, R86333, R86334,
    R86335, R86336, R86337,
    R86338, R86339, R86340,
    R86341, R86342, R86343,
    R86344, R86345, R86346,
    R86347, R86348, R86349,
    R86350, R86351, R86352,
    R86353, R86354, R86355,
    R86356, R86357, R86358,
    R86359, R86360, R86361,
    R86362, R86363, R86364,
    R86365, R86366, R86367,
    R86368, R86369, R86370,
    and R86371 DE4412174
    HIV Inhibitor Peptide Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    GeneSeq Accession the art: HIV protease assays: EP0387231. One can modify the assay to look for
    Y89687 WO9959615 inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    inflammatory disorders, immuno- logic disorders, cancer, viral infections
    HIV Inhibitor Peptide Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    GenSeq Accession the art: HIV protease assays: EP0387231. One can modify the assay to look for
    Y31955 WO9948513 inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    inflammatory disorders, immuno- logic disorders, cancer, viral infections
    HIV Inhibitor Peptide Peptides that inhibit the function/binding of HIV HIV protease activities are known in
    Science 291, 884 (2001); the art: HIV protease assays: EP0387231. One can modify the assay to look for
    Published online 12 Jan. inhibition using any of the disclosed protease inhibitor polypeptides. HIV,
    2001; 10.1126/science.1057453 inflammatory disorders, immuno- logic disorders, cancer, viral infections
    Human monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemoattractant factor processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    hMCP-3 GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession R73915 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9509232 chemokines exert their effects by acting on a family of seven transmembrane G-
    P80098/CCL7_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 7 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 336) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, particularly useful for treating
    bacterial and/or viral menigitis
    Human monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemoattractant factor processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    hMCP-1 GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession R73914 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9509232 chemokines exert their effects by acting on a family of seven transmembrane G-
    P13500/CCL2_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 2 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 337) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, particularly useful for treating
    bacterial and/or viral menigitis
    Human gro-beta Chemokines are a family of related small, secreted proteins involved in biological
    chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accessions R66699 and Members of this family are involved in a similarly diverse range of pathologies
    W17671 WO9429341 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P19875/CXCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-X-C motif chemokine 2 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 338) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, inflammatory dis- orders, blood-
    related disorders, stem cell transplantation, cancer
    Human gro-gamma Chemokines are a family of related small, secreted proteins involved in biological
    chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accessions R66700 and Members of this family are involved in a similarly diverse range of pathologies
    W17672 WO9429341 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P19876/CXCL3_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-X-C motif chemokine 3 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 339) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, inflammatory dis- orders, blood-
    related disorders, stem cell transplantation, cancer
    Human gro-alpha Chemokines are a family of related small, secreted proteins involved in biological
    chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accessions R66698 and Members of this family are involved in a similarly diverse range of pathologies
    W18024 WO9429341 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P09341/GROA_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Growth-regulated alpha protein coupled receptors. Over 40 human chemokines have been described, which
    protein bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 340) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, inflammatory disorders, blood-
    related disorders, stem cell transplantation, cancer
    Human eosinophil- Chemokines are a family of related small, secreted proteins involved in biological
    expressed chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (EEC) GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession W05186 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9632481 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 2 of protein coupled receptors. Over 40 human chemokines have been described, which
    WO9632481 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 401) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, particularly treatment of
    eosinophilia, inflammation, allergies, asthma, leukaemia and lymphoma
    Chemokine-like protein Chemokines are a family of related small, secreted proteins involved in biological
    PF4-414 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R92318 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R99809 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 3C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 402) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and blood- related disorders, particularly
    myelosuppression
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M3 GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    R99812 WO9613587 Members of this family are involved in a similarly diverse range of pathologies
    including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Cancer and blood- related disorders, particularly myelosuppression
    Human interleukin-8 (IL-8) Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    R99814 WO9613587 Members of this family are involved in a similarly diverse range of pathologies
    P10145/IL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Interleukin-8 chemokines exert their effects by acting on a family of seven transmembrane G-
    (isoform 1) protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 341) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Cancer and blood- related disorders, particularly myelosuppression
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M1 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R99815 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R99803 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 4B of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 403) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Cancer and blood- related disorders, particularly myelosuppression
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R99816 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R99805 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 4C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 404) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Cancer and blood- related disorders, particularly myelosuppression
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R99817 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R99806 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 4C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 404) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Cancer and blood- related disorders, particularly myelosuppression.
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R99818 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R99804 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 4C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 404) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Cancer and blood- related disorders, particularly myelosuppression.
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R99819 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R99807 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 4C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 404) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and blood- related disorders, particularly
    myelosuppression.
    Chemokine-like protein IL- Chemokines are a family of related small, secreted proteins involved in biological
    8M8 Full-Length and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Mature GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions R99822 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    R9807 WO9613587 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 4C of WO9613587 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 404) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and blood- related disorders, particularly
    myelosuppression.
    Human foetal spleen expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemo-kine, processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    FSEC GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession R98499 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9622374 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 2 of protein coupled receptors. Over 40 human chemokines have been described, which
    WO9622374 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 405) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders
    Liver expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemokine-1(LVEC-1) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    R95689 WO9616979 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9616979 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 406) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammation of the liver
    Liver expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemokine-2(LVEC-2) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    R95690 WO9616979 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 4 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9616979 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 407) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammation of the liver
    Pituitary expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemokine (PGEC) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    R95691 WO9616979 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 6 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9616979 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 408) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammation, particularly of the liver
    Adenoid-expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemokine (ADEC) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    R97664 WO9617868 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9617868 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 409) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammation, angiogenesis, tumorigenesis,
    musculoskeletal disorders
    Human chemokine CC-2 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W38170 WO9741230 Members of this family are involved in a similarly diverse range of pathologies
    Q16663/CCL15_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 15 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 342) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cell migration, proliferation, and
    differentiation disorders
    Human chemokine HCC-1 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W38171 WO9741230 Members of this family are involved in a similarly diverse range of pathologies
    Q16627/CCL14_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 14 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 343) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cell migration, proliferation, and
    differentiation disorders
    Human chemokine CC- 3 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W38172 WO9741230 Members of this family are involved in a similarly diverse range of pathologies
    Q16627/CCL14_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 14 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 343) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cell migration, proliferation, and
    differentiation
    Novel betachemokine Chemokines are a family of related small, secreted proteins involved in biological
    designated PTEC processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W27271 WO9739126 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9739126 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 410) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, vascular disorders, cancer
    Human CX3C 111 amino Chemokines are a family of related small, secreted proteins involved in biological
    acid chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W23344 Members of this family are involved in a similarly diverse range of pathologies
    WO9727299 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9727299 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 411) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, inflammatory diseases,
    abnormal proliferation, regeneration, degeneration, and atrophy
    Human CCF18 chemokine Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W25942 WO9721812 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 4 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9721812 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 412) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Abnormal physiology and development disorders,
    can also be used as an anti- viral agent
    Human beta- chemokine Chemokines are a family of related small, secreted proteins involved in biological
    H1305 (MCP-2) GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W26655 Members of this family are involved in a similarly diverse range of pathologies
    WO9725427 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P80075/CCL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 8 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 344) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Chemotaxis, blood- related disorders, viral
    infection, HIV, wound healing, cancer
    Human eosinocyte CC Chemokines are a family of related small, secreted proteins involved in biological
    type chemokine eotaxin processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W14990 WO9712914 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P51671/CCL11_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Eotaxin protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 245) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammatory and immune disorders
    Human thymus and Chemokines are a family of related small, secreted proteins involved in biological
    activation regulated processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    cytokine (TARC) GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession W14018 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9711969 chemokines exert their effects by acting on a family of seven transmembrane G-
    Q92583/CCL17_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 17 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 261) using assays known in the art: Methods in molecular Biology, 2000, vol. 138:
    Chemokine Protocols, Edited by A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power
    Humana Press Inc., Totowa, NJ Inflammatory and immune disorders
    Human chemokine beta-8 Chemokines are a family of related small, secreted proteins involved in biological
    short forms GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W16315 Members of this family are involved in a similarly diverse range of pathologies
    WO9712041 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Wildtype chemokine beta- chemokines exert their effects by acting on a family of seven transmembrane G-
    8 provided as: protein coupled receptors. Over 40 human chemokines have been described, which
    P55773|CCL23_HUMAN bind to ~17 receptors thus far identified. Chemokine activities can be determined
    C-C motif chemokine 23 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 459) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, wound healing, immune disorders
    Microphage derived Chemokines are a family of related small, secreted proteins involved in biological
    chemokine, MDC processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W20058 WO9640923 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    O00626/CCL22_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 22 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 345) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammatory diseases, wound healin, angiogenesis
    Human chemokine ZSIG- Chemokines are a family of related small, secreted proteins involved in biological
    35 GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W30565 WO9844117 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 2 of WO including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9844117 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 413) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammatory and immune diseases
    Primate CC chemokine Chemokines are a family of related small, secreted proteins involved in biological
    “ILINCK” GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accesssion W69990 Chemokine activities can be determined using assays known in the art: Methods in
    WO98328658 Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I.
    SEQ ID NO: 4 from Proudfoot, T. N. C. Wells, and C. A. Power. Humana Press Inc., Totowa, NJ.
    WO9832858 Immune and inflammatory disorders, abnormal proliferation, regen- eration,
    (SEQ ID NO: 414) generation and atrophy disorders
    Primate CXC chemokine Chemokines are a family of related small, secreted proteins involved in biological
    “IBICK” GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W69989 Members of this family are involved in a similarly diverse range of pathologies
    WO9832858 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 from chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9832858 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 415) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders, abnormal
    proliferation, regen- eration, generation and atrophy disorders
    Human CC-type Chemokines are a family of related small, secreted proteins involved in biological
    chemokine protein processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    designated SLC Members of this family are involved in a similarly diverse range of pathologies
    (secondary lymphoid including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    chemokine) GeneSeq chemokines exert their effects by acting on a family of seven transmembrane G-
    Accession W69163 protein coupled receptors. Over 40 human chemokines have been described, which
    WO9831809 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    O00585/CCL21_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    C-C motif chemokine 21 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 346) Humana Press Inc., Totowa, NJ. Immune, inflammatory, and infectious disorders,
    cancer
    Human CC chemokine Chemokines are a family of related small, secreted proteins involved in biological
    ELC protein GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W62542 Members of this family are involved in a similarly diverse range of pathologies
    WO9826071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Q99731/CCL19_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 19 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 249) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and infectious diseases, particularly herpes
    virus
    Human DVic-1 C-C Chemokines are a family of related small, secreted proteins involved in biological
    chemokine GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W60649 Members of this family are involved in a similarly diverse range of pathologies
    WO9823750 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9823750 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 416) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Abnormal prolifera- tion, regeneration,
    degeneration, and atrophy disorders, including cancer
    Human C-C chemokine Chemokines are a family of related small, secreted proteins involved in biological
    DGWCC GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W60650 Members of this family are involved in a similarly diverse range of pathologies
    WO9823750 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 6 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9823750 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 417) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cell proliferation disorders,
    cancer
    Human STCP-1 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W62783 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9824907 Members of this family are involved in a similarly diverse range of pathologies
    O00626/CCL22_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 22 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 345) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, particularly T cell related
    disorders, viral infection, and inflammation, especially joint
    Exodus protein GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W61279 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9821330 Members of this family are involved in a similarly diverse range of pathologies
    P78556/CCL20_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 20 chemokines exert their effects by acting on a family of seven transmembrane G-
    (isoform 1) protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 248) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders, angiogenesis,
    cancer, and proliferation disorders, particularly myeloproliferative diseases
    Human Chr19kine protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Acession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W50887 WO9814581 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 10 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9814581 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 418) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and de- generative disorders
    Human T cell mixed Chemokines are a family of related small, secreted proteins involved in biological
    lymphocyte reaction processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    expressed chemokine Members of this family are involved in a similarly diverse range of pathologies
    (TMEC) GeneSeq including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Accession W58703 U.S. chemokines exert their effects by acting on a family of seven transmembrane G-
    Pat. No. 5,780,268 protein coupled receptors. Over 40 human chemokines have been described, which
    SEQ ID NO: 2 of U.S. Pat. bind to ~17 receptors thus far identified. Chemokine activities can be determined
    No. 5,780,268 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 460) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune, inflammatory, and infectious disorders,
    cancer
    Human 6CKine protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W50885 W09814581 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 8 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9814581 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 419) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and de- generative disorders
    human liver and activation Chemokines are a family of related small, secreted proteins involved in biological
    regulated chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (LARC) GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession W57475 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9817800 chemokines exert their effects by acting on a family of seven transmembrane G-
    P78556/CCL20_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 20 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (isoform 1) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 248) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune, inflammatory, and infectious disorders,
    cancer
    RANTES peptide Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W29538 WO9744462 Members of this family are involved in a similarly diverse range of pathologies
    Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    herien as chemokines exert their effects by acting on a family of seven transmembrane G-
    P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 5 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Infectious diseases, particularly HIV
    RANTES 8-68 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W29529 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9744462 Members of this family are involved in a similarly diverse range of pathologies
    Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    herein as chemokines exert their effects by acting on a family of seven transmembrane G-
    P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 5 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Infectious diseases, particularly HIV
    RANTES 9-68 GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession W29528 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9744462 Members of this family are involved in a similarly diverse range of pathologies
    Wildtype Rantes provided including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    herien as chemokines exert their effects by acting on a family of seven transmembrane G-
    P13501/CCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 5 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 241) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Infectious diseases, particularly HIV
    Human chemokine protein Chemokines are a family of related small, secreted proteins involved in biological
    331D5 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W59433 Members of this family are involved in a similarly diverse range of pathologies
    WO9811226 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 12 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9811226 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 420) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Abnormal prolifera- tion, regeneration,
    degeneration, or atrophy, including cancer
    Human chemokine protein Chemokines are a family of related small, secreted proteins involved in biological
    61164 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W59430 Members of this family are involved in a similarly diverse range of pathologies
    WO9811226 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 6 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9811226 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 421) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Abnormal prolifera- tion, regeneration,
    degeneration, or atrophy, including cancer
    Chemokine MCP-4 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W56690 WO9809171 Members of this family are involved in a similarly diverse range of pathologies
    Q99616/CCL13_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 13 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 347) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune, Inflammatory, and infectious diseases
    Human stromal cell- Chemokines are a family of related small, secreted proteins involved in biological
    derived chemokine, SDF-1 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W50766 FR2751658 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P48061/SDF1_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Stromal cell-derived factor 1 protein coupled receptors. Over 40 human chemokines have been described, which
    (isoform beta) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 260) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. HIV infections
    Thymus expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemokine (TECK) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W44397 WO9801557 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    O15444/CCL25_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 25 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 348) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
    Human chemokine MIP- Chemokines are a family of related small, secreted proteins involved in biological
    3alpha GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W44398 Members of this family are involved in a similarly diverse range of pathologies
    WO9801557 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P78556/CCL20_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 20 protein coupled receptors. Over 40 human chemokines have been described, which
    (isoform 1) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 248) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
    Human chemokine MIP- Chemokines are a family of related small, secreted proteins involved in biological
    3beta GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W44399 WO9801557 Members of this family are involved in a similarly diverse range of pathologies
    Q99731/CCL19_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 19 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 249) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
    Human monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemotactic proprotein processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (MCPP) sequence Members of this family are involved in a similarly diverse range of pathologies
    GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    W42072 WO9802459 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 1 of protein coupled receptors. Over 40 human chemokines have been described, which
    WO9802459 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 456) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, respiratory disorders, cancer
    Macrophage- derived Chemokines are a family of related small, secreted proteins involved in biological
    chemokine (MDC) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accessions Members of this family are involved in a similarly diverse range of pathologies
    W40811 and Y24414 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Pat. No. 5,688,927/U.S. chemokines exert their effects by acting on a family of seven transmembrane G-
    Pat. No. 5,932,703 protein coupled receptors. Over 40 human chemokines have been described, which
    O00626/CCL22_HUMAN bind to ~17 receptors thus far identified. Chemokine activities can be determined
    C-C motif chemokine 22 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 345) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune, and inflammatory disorders, cancer
    Macrophage derived Chemokines are a family of related small, secreted proteins involved in biological
    chemokine analogue processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    MDC-eyfy GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession Y24416 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Pat. No. 5,932,703 (“eyfy” chemokines exert their effects by acting on a family of seven transmembrane G-
    disclosed as SEQ ID NO: protein coupled receptors. Over 40 human chemokines have been described, which
    546) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    Wildtype MDC is SEQ ID using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    NO: 2 of 5,932,703 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 422) Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
    Macrophage derived Chemokines are a family of related small, secreted proteins involved in biological
    chemokine analogue MDC processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (n + 1) GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession Y24413 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Pat. No. 5,932,703 chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
    Macrophage derived Chemokines are a family of related small, secreted proteins involved in biological
    chemokine analogue processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    MDC-yl GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession Y24415 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Pat. No. 5,932,703 chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and inflammatory disorders
    Human type CC Chemokines are a family of related small, secreted proteins involved in biological
    chemokine eotaxin
    3 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    protein sequence Members of this family are involved in a similarly diverse range of pathologies
    GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Y43178 JP11243960 chemokines exert their effects by acting on a family of seven transmembrane G-
    Q9Y258/CCL26_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 26 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 349) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Allergic diseases and HIV infection
    Human MCP-3 and human Chemokines are a family of related small, secreted proteins involved in biological
    Muc-1 core epitope (VNT) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    fusion protein GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Acession Y29893 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9946392 chemokines exert their effects by acting on a family of seven transmembrane G-
    Wildtype MCP-3 has the protein coupled receptors. Over 40 human chemokines have been described, which
    sequence: bind to ~17 receptors thus far identified. Chemokine activities can be determined
    P80098/CCL7_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    C-C motif chemokine 7 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 336) Humana Press Inc., Totowa, NJ. Cancer and immune disorders, particularly HIV
    Wildtype Muc-1 has the infection
    sequence:
    P15941|MUC1_HUMAN
    Mucin-1
    (isoform 1)
    (SEQ ID NO: 461)
    Human IP-10 and human Chemokines are a family of related small, secreted proteins involved in biological
    Muc-1 core epitope (VNT) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    fusion protein GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession Y29894 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9946392 chemokines exert their effects by acting on a family of seven transmembrane G-
    Wildtype IP10 has the protein coupled receptors. Over 40 human chemokines have been described, which
    sequence: bind to ~17 receptors thus far identified. Chemokine activities can be determined
    P02778/CXL10_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    C-X-C motif chemokine 10 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 242) Humana Press Inc., Totowa, NJ. Cancer and immune disorders, particularly HIV
    Wildtype Muc-1 has the infection
    sequence:
    P15941|MUC1_HUMAN
    Mucin-1
    (isoform 1)
    (SEQ ID NO: 461)
    Human IP-10 and HIV-1 Chemokines are a family of related small, secreted proteins involved in biological
    gp
    120 hyper- variable processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    region fusion protein Members of this family are involved in a similarly diverse range of pathologies
    GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Y29897 W09946392 chemokines exert their effects by acting on a family of seven transmembrane G-
    Wildtype IP10 has the protein coupled receptors. Over 40 human chemokines have been described, which
    sequence: bind to ~17 receptors thus far identified. Chemokine activities can be determined
    P02778/CXL10_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    C-X-C motif chemokine 10 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 242) Humana Press Inc., Totowa, NJ. Cancer and immune disorders, particularly HIV
    Wildtype gp120 has the infection
    sequence:
    P03378|32-509
    (cleaved product of gp160)
    (SEQ ID NO: 462)
    Human mammary Chemokines are a family of related small, secreted proteins involved in biological
    associated chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (MACK) protein Full- Members of this family are involved in a similarly diverse range of pathologies
    Length and Mature including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    GeneSeq Accessions chemokines exert their effects by acting on a family of seven transmembrane G-
    Y29092 and Y29093 protein coupled receptors. Over 40 human chemokines have been described, which
    WO9936540 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    Full-length: SEQ ID NO: 1 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    of WO9936540 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 423) Humana Press Inc., Totowa, NJ. Breast disease, including cancer
    Mature Form: SEQ ID
    NO: 2 of WO9936540
    (SEQ ID NO: 424)
    Tim-1 protein GeneSeq Chemokines are a family of related small, secreted proteins involved in biological
    Accession Y28290 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    WO9933990 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9933990 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 350) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammation due to stimuli such as heart attacks
    and stroke, infection, physical trauma, UV or ionizing radiation, burns, frostbite or
    corrosive chemicals
    Human Lkn-1 Full-Length Chemokines are a family of related small, secreted proteins involved in biological
    and Mature protein processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accessions Members of this family are involved in a similarly diverse range of pathologies
    Y17280, Y17274, Y17281, including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    and Y17275 WO9928473 chemokines exert their effects by acting on a family of seven transmembrane G-
    and WO9928472 protein coupled receptors. Over 40 human chemokines have been described, which
    Q16663/CCL15_HUMAN bind to ~17 receptors thus far identified. Chemokine activities can be determined
    C-C motif chemokine 15 using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 342) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. HIV infection and cancer, particularly leukemia
    N-terminal modified Chemokines are a family of related small, secreted proteins involved in biological
    chemokine met- hSDF-1 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    alpha GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y05818 WO9920759 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 10 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9920759 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 425) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inhibit or stimulate angiogenesis, inhibit the binding
    of HIV
    N-terminal modified Chemokines are a family of related small, secreted proteins involved in biological
    chemokine met- hSDF-1 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    beta GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y05819 WO9920759 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 11 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9920759 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 426) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inhibit or stimulate angiogenesis, inhibit the binding
    of HIV, antiinflammatory; immunosuppressant
    N-terminal modified Chemokines are a family of related small, secreted proteins involved in biological
    chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GroHEK/hSDF- 1alpha Members of this family are involved in a similarly diverse range of pathologies
    GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Y05820 WO9920759 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 12 of protein coupled receptors. Over 40 human chemokines have been described, which
    WO9920759 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 427) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inhibit or stimulate angiogenesis, inhibit the binding
    of HIV, antiinflammatory; immunosuppressant
    N-terminal modified Chemokines are a family of related small, secreted proteins involved in biological
    chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GroHEK/hSDF- 1beta. Members of this family are involved in a similarly diverse range of pathologies
    GeneSeq Accession including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Y05821 WO9920759 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 13 of protein coupled receptors. Over 40 human chemokines have been described, which
    WO9920759 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 428) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inhibit or stimulate angiogenesis, inhibit the binding
    of HIV, antiinflammatory; immunosuppressant
    Chemokine Eotaxin Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14230 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    P51671/CCL11_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Eotaxin chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 245) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Increase or enhance an inflammatory response, an
    immune response orhaematopoietic cell-associated activity; treat a vascular
    indication; Cancer; enhance wound healing, to prevent or treat asthma, organ
    transplant rejction, rheumatoid arthritis or allergy
    Chemokine hMCP1a Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14225 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound
    healing, cancer, prevent organ transplant rejection, Increase or enhance an
    inflammatory response,
    Chemokine hMCP1b Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14226 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound
    healing, cancer, prevent organ transplant rejection, Increase or enhance an
    inflammatory response,
    Chemokine hSDF1b Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14228 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    P48061/SDF1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Stromal cell-derived factor 1 chemokines exert their effects by acting on a family of seven transmembrane G-
    (isoform beta) protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 260) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound
    healing, cancer, prevent organ transplant rejection, Increase or enhance an
    inflammatory response,
    Chemokine hIL-8 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14229 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    P10145/IL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Interleukin-8 chemokines exert their effects by acting on a family of seven transmembrane G-
    (isoform 1) protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 341) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Immune disorders, Vascular disorders, Wound healing, cancer, prevent organ
    transplant rejection, Increase or enhance an inflammatory response,
    Chemokine hMCP1 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14222 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    P13500/CCL2_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 2 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 337) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound
    healing, cancer, prevent organ transplant rejection, Increase or enhance an
    inflammatory response,
    Chemokine hMCP2 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14223 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    P80075/CCL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 8 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 344) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound
    healing, cancer, prevent organ transplant rejection, Increase or enhance an
    inflammatory response,
    Chemokine hMCP3 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y14224 WO9912968 Members of this family are involved in a similarly diverse range of pathologies
    P80098/CCL7_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 7 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 336) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, Vascular disorders, Wound
    healing, cancer, prevent organ transplant rejection, Increase or enhance an
    inflammatory response,
    C-C chemokine, MCP2 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Y05300 EP905240 Members of this family are involved in a similarly diverse range of pathologies
    P80075/CCL8_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 8 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 344) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammatory, Immune and infectious diseases;
    pulmonary diseases and skin disorders; tumours, and angiogenesis-and
    haematopoiesis- related diseases
    Wild type monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemotactic protein 2 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y07233 EP906954 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P80075/CCL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 8 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 344) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Inflammatory, Immune and infectious diseases;
    pulmonary diseases and skin disorders; tumours, and angiogenesis-and
    haematopoiesis- related diseases
    Truncated monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemotactic protein 2 (6- processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    76) GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y07234 EP906954 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    FIG. 1 of EP905241 and chemokines exert their effects by acting on a family of seven transmembrane G-
    EP906954 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 429) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Inflammatory, Immune and infectious diseases;
    pulmonary diseases and skin disorders; tumours, and angiogenesis-and
    haematopoiesis- related diseases
    Truncated RANTES Chemokines are a family of related small, secreted proteins involved in biological
    protein (3-68) GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accessions Y07236 and Members of this family are involved in a similarly diverse range of pathologies
    Y07232 EP905241; including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    EP906954 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 1 of EP906954 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 430) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Inflammatory, immune and infectious diseases;
    pulmonary diseases and skin disorders; tumours, and angiogenesis-and
    haematopoiesis- related diseases
    Wild type monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemotactic protein 2 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y07237 EP905241 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P80075/CCL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 8 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 344) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Inflammatory, immune and infectious diseases;
    pulmonary diseases and skin disorders; tumours, and angiogenesis-and
    haematopoiesis- related diseases
    Truncated monocyte Chemokines are a family of related small, secreted proteins involved in biological
    chemotactic protein 2 (6-76) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y07238 EP905241 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    FIG. 1 of EP905241 and chemokines exert their effects by acting on a family of seven transmembrane G-
    EP906954 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 429) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Inflammatory, immune and infectious diseases;
    pulmonary diseases and skin disorders; tumours, and angiogenesis-and
    haematopoiesis- related diseases
    A partial CXCR4B protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W97363 EP897980 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    EP897980 chemokines exert their effects by acting on a family of seven transmembrane G-
    (sEQ ID NO: 431) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Soluble CXCR4B receptor polypep- tides may be
    useful for inhibiting chemokine activities and viral infection.
    Interferon gamma- Chemokines are a family of related small, secreted proteins involved in biological
    inducible protein (IP-10) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96709 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    P02778/CXL10_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 10 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 242) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    A monokine induced by Chemokines are a family of related small, secreted proteins involved in biological
    gamma- interferon (MIG) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96710 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    Q07325/CXCL9_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 9 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 351) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Interleukin-8 (IL-8) protein. Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W96711 U.S. Pat. No. Members of this family are involved in a similarly diverse range of pathologies
    5,871,723 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P10145/IL8_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Interleukin-8 protein coupled receptors. Over 40 human chemokines have been described, which
    (isoform 1) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 341) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ; and Holmes et al (1991) Science 253, 1278-80.
    Angiogenesis, Cancer, Inflammatory and Immune disorders, Cardio- Vascular
    disorders, Musco-skeletal disorders
    Epithelial neutrophil Chemokines are a family of related small, secreted proteins involved in biological
    activating protein-78 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (ENA-78) GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession W96712 U.S. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Pat. No. 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    P42830/CXCL5_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 5 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 352) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Growth related oncogene- Chemokines are a family of related small, secreted proteins involved in biological
    alpha (GRO-alpha). processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96713 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    P09341/GROA_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    Growth-regulated alpha bind to ~17 receptors thus far identified. Chemokine activities can be determined
    protein using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 340) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power,
    Humana Press Inc., Totowa, NJ Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Growth related oncogene- Chemokines are a family of related small, secreted proteins involved in biological
    beta (GRO-beta). processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96714 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    P19875/CXCL2_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 2 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 338) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Growth related oncogene- Chemokines are a family of related small, secreted proteins involved in biological
    gamma (GRO-gamma) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96715 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    P19876/CXCL3_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 3 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 339) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    A platelet basic protein Chemokines are a family of related small, secreted proteins involved in biological
    (PBP) GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W96716 U.S. Members of this family are involved in a similarly diverse range of pathologies
    Pat. No. 5,871,723 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P02775/CXCL7_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Platelet basic protein protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 353) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Connective tissue Chemokines are a family of related small, secreted proteins involved in biological
    activating protein-III processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (CTAP-III) GeneSeqAccession Members of this family are involved in a similarly diverse range of pathologies
    S96717 U.S. Pat. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    No. 5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 9 of U.S. protein coupled receptors. Over 40 human chemokines have been described, which
    Pat. No. 5,871,723 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 354) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Beta-thrombo- globulin Chemokines are a family of related small, secreted proteins involved in biological
    protein (beta-TG) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96718 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 10 of U.S. protein coupled receptors. Over 40 human chemokines have been described, which
    Pat. No. 5,871,723 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 355) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Neutrophil activating Chemokines are a family of related small, secreted proteins involved in biological
    peptide-2 (NAP-2) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96719 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    SEQ ID NO: 11 of U.S. protein coupled receptors. Over 40 human chemokines have been described, which
    Pat. No. 5,871,723 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 356) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Granulocyte chemotactic Chemokines are a family of related small, secreted proteins involved in biological
    protein-2 (GCP-2) processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W96720 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    5,871,723 chemokines exert their effects by acting on a family of seven transmembrane G-
    P80162/CXCL6_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 6 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 357) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Angiogenesis, Cancer, Inflammatory and Immune
    disorders, Cardio- Vascular disorders, Musco-skeletal disorders
    Human chemokine MIG- Chemokines are a family of related small, secreted proteins involved in biological
    beta protein GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession W90124 Members of this family are involved in a similarly diverse range of pathologies
    EP887409 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    (SEQ ID NO: 463) chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, viral, parasitic, fungal or
    bacterial infections, Cancer; autoimmune diseases or transplant rejection
    Human ZCHEMO-8 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W82716 WO9854326 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 2 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 432) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders,
    autoimmune disorders and immunodeficiencies, Inflammatory and infectious
    diseases, Vascular disorders, wound healing
    Human Act-2 protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W82717 WO9854326 Members of this family are involved in a similarly diverse range of pathologies
    P13236/CCL4_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 4 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 358) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders,
    autoimmune disorders and immunodeficiencies, Inflammatory and infectious
    diseases, Vascular disorders, wound healing
    Human SISD protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Acession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W82720 WO9854326 Members of this family are involved in a similarly diverse range of pathologies
    P13501/CCL5_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 5 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 241) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders,
    autoimmune disorders and immunodeficiencies, Inflammatory and infectious
    diseases, Vascular disorders, wound healing
    Human MI10 protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W82721 WO9854326 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 37 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 433) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders,
    autoimmune disorders and immunodeficiencies, Inflammatory and infectious
    diseases, Vascular disorders, wound healing
    Human MI1A protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W82722 W09854326 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 38 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 434) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders,
    autoimmune disorders and immunodeficiencies, Inflammatory and infectious
    diseases, Vascular disorders, wound healing
    Human CCC3 protein Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    W82723 WO9854326 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 39 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9854326 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 435) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune disorders, cancer, myelopoietic disorders,
    autoimmune disorders and immunodeficiencies, Inflammatory and infectious
    diseases, Vascular disorders, wound healing
    A human L105 chemokine Chemokines are a family of related small, secreted proteins involved in biological
    designated huL105_3. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W87588 WO9856818 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9856818 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 436) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, wound healing
    A human L105 chemokine Chemokines are a family of related small, secreted proteins involved in biological
    designated huL105_7. processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    W87589 WO9856818 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 4 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9856818 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 437) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, wound healing
    Human mature gro-alpha Chemokines are a family of related small, secreted proteins involved in biological
    polypeptide used to treat processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    sepsis GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession W81498 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9848828 chemokines exert their effects by acting on a family of seven transmembrane G-
    P09341/GROA_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    Growth-regulated alpha bind to ~17 receptors thus far identified. Chemokine activities can be determined
    protein using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 340) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Infectious diseases, sepsis
    Human mature gro- Chemokines are a family of related small, secreted proteins involved in biological
    gamma polypeptide used processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    to treat sepsis GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession W81500 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO9848828 chemokines exert their effects by acting on a family of seven transmembrane G-
    P19876/CXCL3_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-X-C motif chemokine 3 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 339) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Infectious diseases, sepsis
    Human thymus expressed Chemokines are a family of related small, secreted proteins involved in biological
    chemokine TECK and processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    TECK variant GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accessions B19607 and including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    B19608 WO0053635 chemokines exert their effects by acting on a family of seven transmembrane G-
    Wildtype TECK provided protein coupled receptors. Over 40 human chemokines have been described, which
    as: bind to ~17 receptors thus far identified. Chemokine activities can be determined
    O15444/CCL25_HUMAN using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    C-C motif chemokine 25 Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    (SEQ ID NO: 348) Humana Press Inc., Totowa, NJ. Inflammatory disorders, cancer, Immune and
    vascular disorders
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    SDF1alpha GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession B15791 Members of this family are involved in a similarly diverse range of pathologies
    WO0042071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P48061-2/SDF1_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Isoform Alpha of Stromal protein coupled receptors. Over 40 human chemokines have been described, which
    cell-derived factor 1 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (isoform alpha) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 259) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine GRO- Chemokines are a family of related small, secreted proteins involved in biological
    alpha GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15793 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P09341/GROA_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Growth-regulated alpha chemokines exert their effects by acting on a family of seven transmembrane G-
    protein protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 340) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine eotaxin Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15794 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P51671/CCL11_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Eotaxin chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 245) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine MIG Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15803 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    Q07325/CXCL9_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-X-C motif chemokine 9 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 351) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine PF4 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15804 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P02776/PLF4_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Platelet factor 4 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 359) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine I-309 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15805 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P22362/CCL1_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 1 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 360) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine HCC-1 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15806 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    Q16627/CCL14_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C motif chemokine 14 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 361) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine C10 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15807 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    SEQ ID NO: 49 of including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO0042071 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 438) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine CCR-2 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15808 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P41597/CCR2_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-C chemokine receptor chemokines exert their effects by acting on a family of seven transmembrane G-
    type 2 protein coupled receptors. Over 40 human chemokines have been described, which
    (isoform A) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 362) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine ENA- Chemokines are a family of related small, secreted proteins involved in biological
    78 GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15809 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P42830/CXCL5_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-X-C motif chemokine 5 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 352) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    GRObeta GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession B15810 Members of this family are involved in a similarly diverse range of pathologies
    WO0042071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P19875/CXCL2_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-X-C motif chemokine 2 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 338) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine IP-10 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15811 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P02778/CXL10_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    C-X-C motif chemokine 10 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 242) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    SDF1beta GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession B15812 Members of this family are involved in a similarly diverse range of pathologies
    WO0042071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P48061/SDF1_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    Stromal cell-derived factor 1 protein coupled receptors. Over 40 human chemokines have been described, which
    (isoform beta) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 260) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine GRO Chemokines are a family of related small, secreted proteins involved in biological
    alpha GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B15813 WO0042071 Members of this family are involved in a similarly diverse range of pathologies
    P09341/GROA_HUMAN including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Growth-regulated alpha chemokines exert their effects by acting on a family of seven transmembrane G-
    protein protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 340) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    MIP1beta GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession B15831 Members of this family are involved in a similarly diverse range of pathologies
    WO0042071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P13236/CCL4_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 4 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 358) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Autoimmune disorders, Immune, Vascular and
    Inflammatory disorders
    A human C-C chemokine Chemokines are a family of related small, secreted proteins involved in biological
    designated exodus processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    B07939 U.S. Pat. No. including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    6,096,300 chemokines exert their effects by acting on a family of seven transmembrane G-
    P78556/CCL20_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 20 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (isoform 1) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    (SEQ ID NO: 248) Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    L105_7 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession Y96922 U.S. Members of this family are involved in a similarly diverse range of pathologies
    Pat. No. 6,084,071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 4 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9856818 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 437) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Chemotaxis, Gene Therapy, Wound healing
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    L105_3 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession Y96923 U.S. Members of this family are involved in a similarly diverse range of pathologies
    Pat. No. 6,084,071 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    SEQ ID NO: 2 of chemokines exert their effects by acting on a family of seven transmembrane G-
    WO9856818 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 436) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Chemotaxis, Gene Therapy, Wound healing
    Human secondary Chemokines are a family of related small, secreted proteins involved in biological
    lymphoid chemokine processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    (SLC) GeneSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession B01434 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO0038706 chemokines exert their effects by acting on a family of seven transmembrane G-
    O00585/CCL21_HUMAN protein coupled receptors. Over 40 human chemokines have been described, which
    C-C motif chemokine 21 bind to ~17 receptors thus far identified. Chemokine activities can be determined
    (SEQ ID NO: 346) using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, Vascular and Immune disorders
    Human non- ELR CXC Chemokines are a family of related small, secreted proteins involved in biological
    chemokine H174 processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y96310 WO0029439 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    O14625/CXL11_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-X-C motif chemokine 11 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 363) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and Inflammatory disorders, Cancer,
    Haemostatic and thrombolytic activity
    Human non-ELR CXC Chemokines are a family of related small, secreted proteins involved in biological
    chemokine IP10 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession Y96311 Members of this family are involved in a similarly diverse range of pathologies
    WO0029439 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P02778/CXL10_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-X-C motif chemokine 10 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 242) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and Inflammatory disorders, Cancer,
    haemostatic and thrombolytic activity
    Human non-ELR CXC Chemokines are a family of related small, secreted proteins involved in biological
    chemokine Mig GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession Y96313 Members of this family are involved in a similarly diverse range of pathologies
    WO0029439 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    Q07325/CXCL9_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-X-C motif chemokine 9 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 351) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Immune and Inflammatory disorders, Cancer,
    haemostatic and thrombolytic activity
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    Ckbeta-7 GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession Y96280 Members of this family are involved in a similarly diverse range of pathologies
    WO0028035 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    FIG. 1 of WO0028035 chemokines exert their effects by acting on a family of seven transmembrane G-
    (SEQ ID NO: 439) protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, wound healing, inflammatory and
    immunoregulatory disorders
    Human chemokine MIP- Chemokines are a family of related small, secreted proteins involved in biological
    1alpha GeneSeq processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    Accession Y96281 Members of this family are involved in a similarly diverse range of pathologies
    WO0028035 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    P10147/CCL3_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 3 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 364) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, wound healing, inflammatory and
    immunoregulatory disorders
    Human mature chemokine Chemokines are a family of related small, secreted proteins involved in biological
    Ckbeta-7 (optionally processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    truncated) GenSeq Members of this family are involved in a similarly diverse range of pathologies
    Accession Y96282 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    WO0028035 chemokines exert their effects by acting on a family of seven transmembrane G-
    FIG. 1 of WO0028035 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 440) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, wound healing, inflammatory and
    immunoregulatory disorders
    Human chemokine Chemokines are a family of related small, secreted proteins involved in biological
    receptor CXCR3 GeneSeq processes ranging from Chemokine activities can be determined using assays known
    Accession Y79372 in the art: Methods in Molecular Biology, 2000, vol. 138: Soluble CXCR3 polypeptides
    WO0018431 may be useful for inhibiting
    P49682|CXCR3_HUMAN
    C-X-C chemokine receptor
    type 3
    (isoform 1)
    (SEQ ID NO: 240)
    Human neurotactin hematopoiesis, angiogenesis, and leukocyte trafficking. Members of this family are
    chemokine like domain involved in a similarly diverse range of pathologies including inflammation, allergy,
    GeneSeq Accession tissue rejection, viral infection, and tumor biology. The chemokines exert their effects
    Y53259 U.S. Pat. No. by acting on a family of seven transmembrane G-protein coupled receptors. Over 40
    6,043,086 human chemokines have been described, which bind to ~17 receptors thus far
    P78423/X3CL1_HUMAN identified. Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Fractalkine Humana Press Inc., Totowa, NJ. chemokine activities and viral infection.
    (SEQ ID NO: 244)
    Human CC type Chemokines are a family of related small, secreted proteins involved in biological
    chemokine interleukin C processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    GeneSeq Accession Members of this family are involved in a similarly diverse range of pathologies
    Y57771 JP11302298 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    chemokines exert their effects by acting on a family of seven transmembrane G-
    protein coupled receptors. Over 40 human chemokines have been described, which
    bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer and infectious diseases
    Human CKbeta-9 Chemokines are a family of related small, secreted proteins involved in biological
    GeneSeq Accession processes ranging from hematopoiesis, angiogenesis, and leukocyte trafficking.
    B50860 U.S. Pat. No. Members of this family are involved in a similarly diverse range of pathologies
    6,153,441 including inflammation, allergy, tissue rejection, viral infection, and tumor biology. The
    O00585/CCL21_HUMAN chemokines exert their effects by acting on a family of seven transmembrane G-
    C-C motif chemokine 21 protein coupled receptors. Over 40 human chemokines have been described, which
    (SEQ ID NO: 346) bind to ~17 receptors thus far identified. Chemokine activities can be determined
    using assays known in the art: Methods in Molecular Biology, 2000, vol. 138:
    Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
    Humana Press Inc., Totowa, NJ. Cancer, Auto- immune and inflammatory disorders,
    Cardiovascular disorders
    Preproapolipo- protein Apoa-1 participates in the reverse transport of cholesterol from tissues to the liver for
    “paris” variant GeneSeq excretion by promoting cholesterol efflux from tissues and by acting as a cofactor for
    Accession W08602 the lecithin cholesterol acyltransferase (Icat). Lipid binding activity can be determined
    WO9637608 using assays known in the art, such as, for example, the Cholesterol Efflux Assays of
    (SEQ ID NO: 466) Takahaski et al., P.N.A.S., Vol. 96, Issue 20, 11358-11363, Sep. 28, 1999. Useful
    for cardio- vascular disorders, cholesterol disorders, and Hyperlipidaemia
    Preproapolipo- protein Apoa-1 participates in the reverse transport of cholesterol from tissues to the liver for
    “milano” variant 5,721,114 excretion by promoting cholesterol efflux from tissues and by acting as a cofactor for
    SEQ ID NO: 6 of U.S. the lecithin cholesterol acyltransferase (Icat). Lipid binding activity can be determined
    Pat. No. 5,721,114 using assays known in the art, such as, for example, the Cholesterol Efflux Assays of
    (SEQ ID NO: 441) Takahaski et al., P.N.A.S., Vol. 96, Issue 20, 11358-11363, Sep. 28, 1999. Useful
    for cardio- vascular disorders, cholesterol disorders, and Hyperlipidaemia
    Glycodelin-A; Naturally produced female contraceptive that is removed rapidly from the body
    Progesterone- associated following 2-3 days production. Uses include contraception Glycodelin-A activity can
    endometrial protein be determined using the hemizona assay as described in Oehninger, S., Coddington, C. C.,
    GeneSeq Accession Hodgen, G. D., and Seppala, M (1995) Fertil. Steril. 63, 377-383. Naturally
    W00289 WO9628169 derived contraceptive useful for the prevention of pregnancy.
    P09466/PAEP_HUMAN
    Glycodelin
    (SEQ ID NO: 365)
    NOGO-A Genbank NOGO polypeptides are potent inhibitors of neurite growth. Inhibition of Neurite
    Accession CAB99248 outgrowth. Antagonists to NOGO polypeptides may promote the outgrowth of
    (SEQ ID NO: 366) neurites, thus inducing regeneration of neurons. NOGO-A polypep- tide antagonists
    are useful for the pro- motion of neural growth, which could be useful in the treatment
    of neural disorders and dys- function due to de- generative diseases or trauma; useful
    in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons
    or to promote the structural plasticity of the CNS.
    NOGO-B Genbank NOGO polypeptides are potent inhibitors of neurite growth. Inhibition of Neurite
    Accession CAB99249 outgrowth. Antagonists to NOGO polypeptides may promote the outgrowth of
    (SEQ ID NO: 367) neurites, thus inducing regeneration of neurons. NOGO-B polypep- tide antagonists
    are useful for the pro- motion of neural growth, which could be useful in the treatment
    of neural disorders and dys- function due to de- generative diseases or trauma; useful
    in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons
    or to promote the structural plasticity of the CNS.
    NOGO-C Genbank NOGO polypeptides are potent inhibitors of neurite growth. Inhibition of Neurite
    Accession CAB99250 outgrowth. Antagonists to NOGO polypeptides may promote the outgrowth of
    (SEQ ID NO: 368) neurites, thus inducing regeneration of neurons. NOGO-C polypep- tide antagonists
    are useful for the pro- motion of neural growth, which could be useful in the treatment
    of neural disorders and dys- function due to de- generative diseases or trauma; useful
    in the treatment of neo- plastic diseases of the CNS; induce regeneration of neurons
    or to promote the structural plasticity of the CNS.
    NOGO-66 Receptor NOGO polypeptides are potent inhibitors of neurite growth, and are thought to
    Genbank Accession mediate their effects through the NOGO-66 Receptor. Inhibition of Neurite outgrowth
    AAG53612 by mediating the biological effects of NOGO polypeptides. Soluble NOGO- 66
    (SEQ ID NO: 369) receptor polypeptides may promote the outgrowth of neurites, thus inducing
    regeneration of neurons. NOGO-66 receptor polypeptides are useful for the
    promotion of neural growth, which could be useful in the treatment of neural disorders
    and dys- function due to de- generative diseases or trauma; useful in the treatment of
    neo- plastic diseases of the CNS; induce regeneration of neurons or to promote the
    structural plasticity of the CNS.
    Antibodies specific for These antibodies are useful for the promotion of neurite outgrowth Collapsin activity,
    collapsin U.S. Pat. No. which is thought to inhibit the outgrowth of neurites, can be assayed in the presence
    5,416,197 of antibodies specific for collapsing using assays known in the art, such as, for
    Wildtype collapsin has the example, the collapse assay disclosed by Luo et al., Cell 1993 Oct. 22; 75(2): 217-27
    sequence: Useful for the pro- motion of neural growth, which could be useful in the treatment of
    SEQ ID NO: 2 of 5,416,197 neural disorders and dys- function due to de- generative diseases or trauma.
    (SEQ ID NO: 464)
    Humanized Anti-VEGF These agents have anti-inflammatory and anti-cancer applications VEGF activity can
    Antibodies, and fragments be determined using assays known in the art, such as those disclosed in International
    thereof WO9845331 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    cells, such as vascular endothelial cells. Antagonists may be useful as anti-
    angiogenic agents, and may be applicable for cancer
    Humanized Anti-VEGF These agents have anti-inflammatory and anti-cancer applications VEGF activity can
    Antibodies, and fragments be determined using assays known in the art, such as those disclosed in International
    thereof WO0029584 Publication No. WO0045835, for example. Promotion of growth and proliferation of
    cells, such as vascular endothelial cells. Hematopoietic and immune dis- orders.
    Antagonists may be useful as anti-angiogenic agents, and may be applicable for
    cancer
    Membrane bound proteins Cancer, Immune Disorders These proteins can be used for linking bioactive
    GeneSeq. Accession molecules to cells and for modulating biological activities of cells, using the
    Y66631-Y66765 polypeptides for specific targeting. The polypeptide targeting can be used to kill the
    WO9963088 target cells, e.g. for the treatment of cancers. These proteins are useful for the
    treatment of immune system disorders. Activities can be determined using assay
    known in the art, such as, for example, the assays disclosed in International
    Publication No. WO0121658.
    Secreted and Cancer, Immune Disorders These proteins can be used for linking bioactive
    Transmembrane molecules to cells and for modulating biological activities of cells, using the
    polypeptides GeneSeq polypeptides for specific targeting. The polypeptide targeting can be used to kill the
    Accession B44241-B44334 target cells, e.g. for the treatment of cancers. These proteins are useful for the
    WO0053756 treatment of immune system disorders. Activities can be determined using assay
    known in the art, such as, for example, the assays disclosed in International
    Publication No. WO0121658.
    Secreted and Cancer, Immune Disorders These proteins can be used for linking bioactive
    Transmembrane molecules to cells and for modulating biological activities of cells, using the
    polypeptides GeneSeq polypeptides for specific targeting. The polypeptide targeting can be used to kill the
    Accession Y41685-Y41774 target cells, e.g. for the treatment of cancers. These proteins are useful for the
    WO9946281 treatment of immune system disorders. Activities can be determined using assay
    known in the art, such as, for example, the assays disclosed in International
    Publication No. WO0121658.
    Interleukin 2 (IL-2) Metabolic Disease, Type 1 diabetes, Graft-versus-host disease.
    SEQ ID NO: 548
    Interleukin 15_vA Obesity, Metabolic Disease, Diabetes.
    (IL-15_vA)
    SEQ ID NO: 549
    Interleukin 15_vB Obesity, Metabolic Disease, Diabetes.
    (IL-15_vB)
    SEQ ID NO: 550
    Interleukin 15_vC Obesity, Metabolic Disease, Diabetes.
    (IL-15_vC)
    SEQ ID NO: 551
    Interleukin 15_vD Obesity, Metabolic Disease, Diabetes.
    (IL15_vD)
    SEQ ID NO: 552
    Interleukin 15_vE Obesity, Metabolic Disease, Diabetes.
    (IL15_vE)
    SEQ ID NO: 553
    Interleukin 15_vF Obesity, Metabolic Disease, Diabetes.
    (IL15_vF)
    SEQ ID NO: 565
    Interleukin 22 Metabolic Disease, Diabetic Ulcers, Inflamatory Bowel Diseases.
    (IL22)
    SEQ ID NO: 554
    Fibroblast Growth Factor 1 Diabetes, Metabolic Disease, Obesity.
    (FGF1)
    SEQ ID NO: 555
    Fibroblast Growth Factor Diabetes, Metabolic Disease, Obesity.
    1_vA See Nature 513, 436-439 (18 Sep. 2014) doi: 10.1038/nature13540.
    (FGF1_vA)
    SEQ ID NO: 556
    Fibroblast Growth Factor Diabetes, Metabolic Disease, Obesity.
    1_vB See Proc Natl Acad Sci USA. 1991 Apr. 1; 88(7): 2893-2897.
    (FGF1_vB)
    SEQ ID NO: 557
    Fibroblast Growth Factor Diabetes, Metabolic Disease, Obesity.
    1_vC
    (FGF1_vC)
    SEQ ID NO: 566
    Fibroblast Growth Factor Chronic liver disease, primary biliary cirrhosis, bile acid-induced liver damage.
    19_vA See Cancer Res. 2014 Jun. 15; 74(12): 3306-16. doi: 10.1158/0008-5472.CAN-14-
    (FGF19_vA) 0208. Epub 2014 Apr. 11. Regulates bile acid metabolism without tumorigenicity.
    SEQ ID NO: 558
    Fibroblast Growth Factor Metabolic Disease, Fibrotic Diseases.
    21
    (FGF21)
    SEQ ID NO: 559
    Fibroblast Growth Factor Hyperphosphatemic familial tumoral calcinosis.
    23
    (FGF23)
    SEQ ID NO: 560
    Brain-Derived Neurological diseases (including Alzheimer's Disease, Autism, Huntington's Disease,
    Neurotrophic Factor Parkinson's Disease, and Depression), obesity, metabolic disease.
    (BDNF)
    SEQ ID NO: 561
    Serpin Family A Member 1 alpha-1-antitrypsin deficiency.
    (SERPINA1)
    SEQ ID NO: 584
    SEQ ID NO: 585
    Serpin Peptidase Inhibitor, Diabetes.
    Clade B (Ovalbumin),
    Member 1
    (SERPINB1)
    SEQ ID NO: 562
    CASPASE1 Diabetes.
    SEQ ID NO: 563
    Leukemia Inhibitory Factor Muscular dystrophy, atherosclerosis, kidney disease
    (LIF)
    SEQ ID NO: 564
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 1 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK1) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 567 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 2 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK2) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 568 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 3 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK3) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 569 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 3 Sol hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK3_SOL) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 570 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 4 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK4) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 571 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 5 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK5) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 572 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 6 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK6) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 573 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK7) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 574 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 8 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK8) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 575 peptide
    Proprotein Convertase Cardiovascular disease, hypercholesterolemia, heterozygous familial
    Subtilisin/Kexin Type 9 hypercholesterolemia (HeFH), atherosclerotic cardiovascular disease such as heart
    (PCSK9) attacks or strokes, increasing the amount of a functional protein, polypeptide or
    SEQ ID NO: 576 peptide
    Membrane-Bound IFAP syndrome, increasing the amount of a functional protein, polypeptide or peptide
    Transcription Factor
    Peptidase, Site 2
    (MBTPS2)
    SEQ ID NO: 577
    Carboxypeptidase E Endocrine disorders, such as, for example, obesity and infertility;
    (CPE) hyperproinsulinemia; metabolic syndrome, increasing the amount of a functional
    SEQ ID NO: 578 protein, polypeptide or peptide
  • In various embodiments, the nucleic acid drug, including synthetic RNA, is administered is a manner that it effects one or more of keratinocytes and fibroblasts (e.g. causes these cells to express one or more therapeutic proteins). For example, present methods allow for methods in which a patient's cells are used to generate a therapeutic protein and the levels of such protein are tailored by synthetic RNA dosing.
  • In a specific embodiment, the synthetic RNA targets a soluble protein. In some embodiments, the synthetic RNA targets a protein of one or more of the following families of proteins: transforming growth factor (TGF) beta, bone morphogenetic proteins (BMPs), Fibroblast growth factors (FGFs), vascular endothelial growth factors (VEGFs), and interleukins. The terms “family”, “superfamily”, and “subfamily” can be used interchangeably
  • In a specific embodiment, the synthetic RNA targets a member of the TGF beta family. TGF-β superfamily proteins comprise cytokines characterized by six-conserved cysteine residues (Lander et al., (2001) Nature, 409:860-921). The human genome contains at least about 42 open reading frames encoding TGF-β superfamily proteins. TGF-β superfamily proteins can at least be divided into the BMP subfamily and the TGF-β subfamily based on sequence similarity and the specific signaling pathways that they activate. In various embodiments, the synthetic RNA targets one or more of TGFs (e.g., TGF-β1, TGF-β2, and TGF-β3), activins (e.g., activin A) and inhibins, macrophage inhibitory cytokine-1 (MIC-1), Mullerian inhibiting substance, anti-Mullerian hormone, and glial cell line derived neurotrophic factor (GDNF).
  • The TGF-β superfamily comprises a subset of the cysteine knot cytokine superfamily. Additional members of the cysteine knot cytokine superfamily include, but are not limited to, platelet derived growth factor (PDGF), vascular endothelial growth factor (VEGF), placenta growth factor (P1GF), Noggin, neurotrophins (BDNF, NT3, NT4, and PNGF), gonadotropin, follitropin, lutropin, interleukin-17, and coagulogen. This family of proteins is also among the targets encompassed by the present invention.
  • In various embodiments, the present invention relates to targeting a TGF beta family member for treatment or prevention of various immunological disorders, cancer, bronchial asthma, lung fibrosis, heart disease, diabetes, hereditary hemorrhagic telangiectasia, Marfan syndrome, Vascular Ehlers-Danlos syndrome, Loeys-Dietz syndrome, Parkinson's disease, chronic kidney disease, multiple sclerosis and AIDS.
  • In a specific embodiment, the synthetic RNA targets a member of the BMP family. The BMP subfamily includes, but is not limited to, BMP-2, BMP-3 (osteogenin), BMP-3b (GDF-10), BMP-4 (BMP-2b), BMP-5, BMP-6, BMP-7 (osteogenic protein-1 or OP-1), BMP-8 (OP-2), BMP-8B (OP-3), BMP-9 (GDF-2), BMP-10, BMP-11 (GDF-11), BMP-12 (GDF-7), BMP-13 (GDF-6, CDMP-2), BMP-15 (GDF-9), BMP-16, GDF-1, GDF-3, GDF-5 (CDMP-1), and GDF-8 (myostatin). In various embodiments, the synthetic RNA targets one or more of BMP-2, BMP-3 (osteogenin), BMP-3b (GDF-10), BMP-4 (BMP-2b), BMP-5, BMP-6, BMP-7 (osteogenic protein-1 or OP-1), BMP-8 (OP-2), BMP-8B (OP-3), BMP-9 (GDF-2), BMP-10, BMP-11 (GDF-11), BMP-12 (GDF-7), BMP-13 (GDF-6, CDMP-2), BMP-15 (GDF-9), BMP-16, GDF-1, GDF-3, GDF-5 (CDMP-1), and GDF-8 (myostatin). BMPs are sometimes referred to as Osteogenic Protein (OPs), Growth Differentiation Factors (GDFs), or Cartilage-Derived Morphogenetic Proteins (CDMPs). In a specific embodiment, the synthetic RNA targets one or more BMP fusions (e.g. as described in US Patent Publication No. 2009/0202638, the entire contents of which are hereby incorporated by reference) and/or one or more BMP mutants (e.g. as described in US Patent Publication No. 2011/0039773, the entire contents of which are hereby incorporated by reference).
  • In various embodiments, the present invention relates to targeting a BMP family member for regenerative medicine or metabolic applications, including without limitation orthopedic applications such as spinal fusions, nonunions and oral surgerie, metabolic disease, prediabetes, diabetes, thermogenesis, insulin sensitivity, insulin resistance, and adipogenesis, including brown-fat adipogenesis. Various other metabolic applications are described elesewhere herein. In various embodiments, the present invention relates to targeting a BMP family member, including BMP-7, for treatment of chronic kidney disease (CKD) and/or to reverse the loss of glomeruli due to sclerosis.
  • In various embodiments, the present invention relates to targeting a BMP family member to induce proliferation of bone and cartilage in a variety of locations in the body. For example, repair of joints such as knee, elbow, ankle, and finger joints are contemplated by the invention. For example, targeting a BMP family member may result in regenerating cartilage in patients suffering from arthritis or other cartilage degenerating diseases. Further, the invention pertains to treating tears in cartilage due to injury. In addition, the invention is useful for inducing bone growth in patients, for instance, by way of non-limitation, for use in treating patients suffering from bone fractures or breaks, osteoporosis, or patients in need of spinal fusion or for repair of the spine, vertebrae or the like.
  • In various embodiments, the present invention relates to targeting a BMP family member to induce a developmental cascade of bone morphogenesis and tissue morphogenesis for a variety of tissues in mammals different from bone or bone cartilage. This morphogenic activity includes the ability to induce proliferation and differentiation of progenitor cells, and the ability to support and maintain the differentiated phenotype through the progression of events that results in the formation of bone, cartilage, non-mineralized skeletal or connective tissues, and other adult tissues.
  • For example, the present invention may be used for treatment to prevent loss of and/or increase bone mass in metabolic bone diseases. General methods for treatment to prevent loss of and/or increase bone mass in metabolic bone diseases using osteogenic proteins are disclosed in U.S. Pat. No. 5,674,844, the entire contents of which are hereby incorporated by reference. Further the present compositions and methods find use in replacing or repairing bone or cartilage at injury sites such as bone breaks, bone fractures, and cartilage tears, periodontal tissue regeneration (e.g. general methods for periodontal tissue regeneration using osteogenic proteins are disclosed in U.S. Pat. No. 5,733,878, the entire contents of which are hereby incorporated by reference), liver regeneration, including following a partial hepatectomy (see, e.g., U.S. Pat. No. 5,849,686, the entire contents of which are hereby incorporated by reference), treatment of chronic renal failure (see, e.g., U.S. Pat. No. 6,861,404, the entire contents of which are hereby incorporated by reference), enhancing functional recovery following central nervous system ischemia or trauma (see, e.g. U.S. Pat. No. 6,407,060, the entire contents of which are hereby incorporated by reference), inducing dendritic growth (see, e.g., U.S. Pat. No. 6,949,505, the entire contents of which are hereby incorporated by reference), inducing neural cell adhesion (see, e.g., U.S. Pat. No. 6,800,603, the entire contents of which are hereby incorporated by reference), and treatment and prevention of Parkinson's disease (see, e.g., U.S. Pat. No. 6,506,729, the entire contents of which are hereby incorporated by reference). As another example, the present compositions and methods, including when targeting one or more BMPs, can be used to induce dentinogenesis. To date, the unpredictable response of dental pulp tissue to injury is a basic clinical problem in dentistry. Using standard dental surgical procedures, small areas (e.g., 2 mm) of dental pulps can be surgically exposed by removing the enamel and dentin immediately above the pulp (by drilling) of sample teeth, performing a partial amputation of the coronal pulp tissue, inducing hemostasis, application of the pulp treatment, and sealing and filling the cavity by standard procedures.
  • In various embodiments, the present invention relates to targeting a BMP family member for the treatment of one or more metabolic-related disorders as described herein.
  • In a specific embodiment, the synthetic RNA targets a member of the FGF family. FGFs are a family of growth factors, with members involved in angiogenesis, wound healing, embryonic development and various endocrine signaling pathways. In various embodiments, any one of the following FGFs are targets of the invention: FGF1, FGF2, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, FGF10, FGF11, FGF12, FGF13, FGF14 (FGF11, FGF12, FGF13, and FGF14 being FGF homologous factors 1-4 (FHF1-FHF4)), FGF16, FGF17, FGF18, FGF19 (aka FHF15/19), FGF20, FGF21, FGF22 and FGF22. In some embodiments, the synthetic RNA targets a cognate receptor of any of the above FGFs. In various embodiments, the present invention relates to targeting a FGF family member to a treat or prevent disease or disorder associated with abnormal function and/or expression of an FGF, a metabolic disease or disorder (e.g., diabetes, obesity, dyslipidemia, hyperglycemia, hyperinsulinemia, hypertension, hepatosteaotosis such as non-alcoholic steatohepatitis (NASH) etc.), cancer, a disease or disorder associated with impaired lipid metabolism a disease or disorder associated with impaired renal function, a disease or disorder associated with impaired hepatic function, abnormal cell proliferation, a vascular disease or disorder (e.g., coronary artery disease, peripheral artery disease, atherosclerosis, abdominal aortic aneurysm, a blood clot, deep vein thrombosis, venous stasis disease, phlebitis, varicose veins etc.), angiogenesis, atherosclerosis, a cardiovascular disease or disorder, a disease or disorder associated with impaired blood vessel formation, a disease or disorder associated with impaired cell signaling, a disease or disorder associated with impaired kinase activity, and a disease or disorder associated with impaired uptake of glucose into adipocytes.
  • In a specific embodiment, the synthetic RNA targets a member of the VEGF family. In some embodiments, the target is one or more of VEGF-A (including all isoforms, e.g. VEGF121, VEGF165 and VEGF189), placenta growth factor (PGF, including all isoforms, e.g. PGF-1, PGF-2, and PGF-3), VEGF-B, VEGF-C and VEGF-D, and any variants thereof (see, e.g. U.S. Pat. No. 9,078,860, the entire contents of which are hereby incorporated by reference). In some embodiments, the synthetic RNA targets a cognate receptor of any of the above VEGFs. The present invention also encompasses as targets VEGF-related proteins including orf virus-encoded VEGF-like proteins referred to as VEGF-E and a series of snake venoms referred to as VEGF-F. VEGFs and VEGF-related proteins are members of the Platelet Derived Growth Factor (PDGF) supergene family of cystine knot growth factors. All members of the PDGF supergene family share a high degree of structural homology with PDGF.
  • In various embodiments, the present invention relates to targeting a VEGF family member to treat diseases and conditions associated with angiogenesis, including but not limited to, solid tumor cancers, hemangiomas, rheumatoid arthritis, osteoarthritis, septic arthritis, asthma, atherosclerosis, idiopathic pulmonary fibrosis, vascular restenosis, arteriovenous malformations, meningiomas, neovascular glaucoma, psoriasis, Kaposi's Syndrome, angiofibroma, hemophilic joints, hypertrophic scars, Osler-Weber syndrome, pyogenic granuloma, retrolental fibroplasias, scleroderma, trachoma, von Hippel-Lindau disease, vascular adhesion pathologies, synovitis, dermatitis, neurological degenerative diseases, preeclampsia, unexplained female infertility, endometriosis, unexplained male infertility, pterygium, wounds, sores, skin ulcers, gastric ulcers, and duodenal ulcers. In various embodiments, the present invention relates to targeting a VEGF family member to treat angiogenesis-associated eye diseases, including without limitation any eye disease associated with abnormal intraocular neovascularization, including but not limited to retinopathy of prematurity, diabetic retinopathy, retinal vein occlusion, and age-related macular degeneration, as well diabetic macular edema and retinal vein occlusion. In an embodiment, the present compositions and methods relate to the treatment of wet age-related macular degeneration.
  • In a specific embodiment, the synthetic RNA targets a member of the interleukin family. The interleukins represent a large group of cytokines with diverse functions and were first characterized by expression in leukocytes and have since been shown to be expressed in a wide variety of cells, for example macrophages, TH-1 and TH-2 cells, T-lymphocytes, monocytes and bone marrow stroma. Broadly, the function of the immune system depends in a large part on the expression and function of the interleukins. In some embodiments, the target is one or more of interleukins 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 35 and 36, and, within each species of interleukin, various isotypes and/or interleukin receptors (e.g., IL-1R, IL-2R, IL-3R, IL-4R, IL-5R, IL-6R, IL-7R, IL-8R, IL-9R, IL-10R, IL-11R, IL-12R, IL-13R, IL-14R, IL-15R, IL-16R, IL-17R, IL-18R, IL-19R, IL-20R, IL-21R, IL-22R, IL-23R, IL-24R, IL-25R, IL-26, IL-27R, IL-28R, IL-29R, IL-30R, IL-31R, IL-32R, IL-33R, IL-34R, IL-35R, and IL-36R). In a specific embodiment, both IL-15 and IL-15R (e.g., IL-15RA) are targeted. Without wishing to be bound by theory, it is believed that the targeting of both interleukin (e.g., IL-15) and its cognitive interleukin receptor (e.g., IL-15RA) results in synergistic beneficial effects. In various embodiments, the present invention relates to targeting a member of the interleukin family to treat cancer, inflammatory, respiratory, autoimmune, cardiovascular, neurological, metabolic, and/or proliferative diseases, disorders, and/or conditions in a subject or organism, as described herein. In a specific embodiment, the present invention relates to targeting a member of the interleukin family to treat cancer. In a specific embodiment, the present invention relates to targeting a member of the interleukin family to treat rheumatoid arthritis.
  • In a specific embodiment, the synthetic RNA targets an EPO gene or a derivative thereof (e.g. SEQ ID NO: 164, SEQ ID NO: 165, SEQ ID NO: 166, and SEQ ID NO: 167). Some embodiments are related to NOVEPOETIN protein (SEQ ID NO: 167). Other embodiments are related to NOVECRIT (SEQ ID NO: 168).
  • Erythropoietin can stimulate erythropoiesis in anemic patients with chronic renal failure in whom the endogenous production of erythropoietin is impaired. Without being bound by theory, because of the length of time often required for erythropoiesis (several days for erythroid progenitors to mature and be released into the circulation), a clinically significant increase in hemoglobin is usually not observed in less than two weeks and may require up to ten weeks in some patients. The present methods and compositions provide, in some embodiments, more rapid therapeutic effect. The present methods and compositions provide, in some embodiments, more rapid therapeutic effect, for instance, when compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic. The present methods and compositions provide, in some embodiments, a sustained therapeutic effect. The present methods and compositions provide, in some embodiments, a sustained therapeutic effect, for instance, when compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic.
  • For instance, for EPO, the present methods and compositions provide a clinically significant increase in hematocrit in less than about 6 weeks, or less than about 5 weeks, or less than about 4 weeks, or less than about 3 weeks, or less than about 2 weeks, or less than about 1 week. In some embodiments, the present methods and compositions provide a clinically significant increase in hematocrit in about 2 weeks, or about 10 days, or about 1 week, or about 3 days, or about 1 day. In various embodiments, the present methods and compositions accelerate the process by which erythroid progenitors mature and are released into the circulation.
  • In some embodiments, the present EPO-related compositions find use in decreasing the dose and/or frequency of administration when compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic. For instance, the present EPO-related compositions may find use in treatment regimens for the diseases disclosed herein (including, without limitation, one or more anemias) that involve administration on a monthly, or biweekly, or weekly basis. In some embodiments, therefore, the present EPO-related compositions reduce the need for daily, or, in some embodiments, weekly, administration. In some embodiments, the present EPO-related compositions require lower maintenance doses as compared to wild type EPO and/or EPO without non-canonical nucleotides and/or EPO delivered as a protein biologic. Certain embodiments are particularly useful for treating chemotherapy-induced anemia. Other embodiments are particularly useful for treating anemia associated with inflammation, including, but not limited to, rheumatoid arthritis.
  • In some embodiments, the present the present methods and compositions provide a fast and robust response that obviates the need for RBC transfusion. For instance, in some embodiments, the present methods and compositions allow for treatment of patients do not consent to transfusions.
  • In some embodiments, the present methods and compositions increase the rate of increase in hematocrit. In some embodiments, the present methods and compositions maintain elevated hematocrits (e.g. of 25%, or 30%, or 35%, or 40% or more) for a sustained period (e.g. about 1 month, or about 2 months, or about 3 months, or about 4 months, or about 5 months, or about 6 months, or about 9 months).
  • In some embodiments, the present methods and compositions stimulate red blood cell production. In some embodiments, the present methods and compositions stimulate division and differentiation of committed erythroid progenitors in the bone marrow.
  • In some embodiments, including, without limitation, when targeting EPO, the present invention relates to the treatment of one or more of anemia, including anemia resulting from resulting from chronic kidney disease (e.g. from dialysis) and/or chemotherapy and/or HIV treatment (e.g. Zidovudine (INN) or azidothymidine (AZT)), inflammatory bowel disease (e.g. Crohn's disease and ulcer colitis), anemia linked to inflammatory conditions (e.g. arthritis, lupus, IBD), anemia linked to diabetes, schizophrenia, cerebral malaria, as aplastic anemia, and myelodysplasia from the treatment of cancer (e.g. chemotherapy and/or radiation), and various myelodysplastic syndrome diseases (e.g. sickle cell anemia, hemoglobin SC disease, hemoglobin C disease, alpha- and beta-thalassemias, neonatal anemia after premature birth, and comparable conditions).
  • In some embodiments, including, without limitation, when targeting EPO, the present invention relates to the treatment of, or a patient having one or more of cancer, heart failure, autoimmune disease, sickle cell disease, thalassemia, blood loss, transfusion reaction, diabetes, vitamin B12 deficiency, collagen vascular disease, Shwachman syndrome, thrombocytopenic purpura, Celiac disease, endocrine deficiency state such as hypothyroidism or Addison's disease, autoimmune disease such as Crohn's Disease, systemic lupus erythematosis, rheumatoid arthritis or juvenile rheumatoid arthritis, ulcerative colitis immune disorders such as eosinophilic fasciitis, hypoimmunoglobulinemia, or thymoma/thymic carcinoma, graft vs. host disease, preleukemia, Nonhematologic syndrome (e.g. Down's, Dubowwitz, Seckel), Felty syndrome, hemolytic uremic syndrome, myelodysplasic syndrome, nocturnal paroxysmal hemoglobinuria, osteomyelofibrosis, pancytopenia, pure red-cell aplasia, Schoenlein-Henoch purpura, malaria, protein starvation, menorrhagia, systemic sclerosis, liver cirrhosis, hypometabolic states, congestive heart failure, chronic infections such as HIV/AIDS, tuberculosis, oseomyelitis, hepatitis B, hepatitis C, Epstein-barr virus or parvovirus, T cell leukemia virus, bacterial overgrowth syndrome, fungal or parasitic infections, and/or red cell membrane disorders such as hereditary spherocytosis, hereditary elliptocytosis, heriditray pyrpoikilocytosis, hereditary stomatocytosis, red cell enzyme defects, hypersplenism, immune hemolysis or paroxysmal nocturnal hemoglobinuria.
  • In some embodiments, including, without limitation, when targeting EPO, the present invention relates to the treatment of, or a patient having anemia, i.e. a condition in which the number of red blood cells and/or the amount of hemoglobin found in the red blood cells is below normal. In various embodiments, the anemia may be acute or chronic. For example, the present anemias include but are not limited to iron deficiency anemia, renal anemia, anemia of chronic diseases/inflammation, pernicious anemia such as macrocytic achylic anemia, juvenile pernicious anemia and congenital pernicious anemia, cancer-related anemia, chemotherapy-related anemia, radiotherapy-related anemia, pure red cell aplasia, refractory anemia with excess of blasts, aplastic anemia, X-lined siderobalstic anemia, hemolytic anemia, sickle cell anemia, anemia caused by impaired production of ESA, myelodysplasia syndromes, hypochromic anemia, microcytic anemia, sideroblastic anemia, autoimmune hemolytic anemia, Cooley's anemia, Mediterranean anemia, Diamond Blackfan anemia, Fanconi's anemia and drug-induced immune hemolytic anemia. Anemia may cause serious symptoms, including hypoxia, chronic fatigue, lack of concentration, pale skin, low blood pressure, dizziness and heart failure.
  • In some embodiments, the anemia is induced by chemotherapy is one cause of anemia. For instance, the chemotherapy may be any myelosuppressive chemotherapy. In some embodiments, the chemotherapy is one or more platinum-based drugs including cisplatin (e.g. PLATINOL) and carboplatin (e.g. PARAPLATIN). In some embodiments, the chemotherapy is any one of the agents described herein. In some embodiments, the chemotherapy is any agent described in Groopman et al. J Natl Cancer Inst (1999) 91 (19): 1616-1634, the contents of which are hereby incorporated by reference in their entireties. In some embodiments, the present compositions and methods are used in the treatment of chemotherapy-related anemia in later stage cancer patients (e.g. a stage IV, or stage III, or stage II cancer). In some embodiments, the present compositions and methods are used in the treatment of chemotherapy-related anemia in cancer patients receiving dose-dense chemotherapy or other aggressive chemotherapy regimens.
  • In some embodiments, the present invention relates to the treatment of anemia in a patient having one or more blood-based cancers, such as leukemia, lymphoma, and multiple myeloma. Such cancers may affect the bone marrow directly. Further, the present invention relates to metastatic cancer that has spread to the bone or bone marrow. In some embodiments, the present invention relates to the treatment of anemia in a patient undergoing radiation therapy. Such radiation therapy may damage the bone marrow, lowering its ability to make red blood cells. In further embodiments, the present invention relates to the treatment of anemia in a patient having a reduction or deficiency of one or more of iron, vitamin B12, and folic acid. In further embodiments, the present invention relates to the treatment of anemia in a patient having excessive bleeding including without limitation, after surgery or from a tumor that is causing internal bleeding. In further embodiments, the present invention relates to the treatment of anemia in a patient having anemia of chronic disease.
  • In some embodiments, the present invention relates to the treatment of anemia resulting from chronic renal failure. In some embodiments, the present invention relates to the treatment of anemia resulting from the use of one or more renal replacement therapies, inclusive of dialysis, hemodialysis, peritoneal dialysis, hemofiltration, hemodiafiltration, and renal transplantation.
  • In some embodiments, the present invention relates to the treatment of anemia in patients with chronic kidney disease who are not on dialysis. For instance, the present invention relates to patients in stage 1 CKD, or stage 2 CKD, or stage 3 CKD, or stage 4 CKD, or stage 5 CKD. In some embodiments, the present patient is stage 4 CKD or stage 5 CKD. In some embodiments, the present patient has undergone a kidney transplant. In some embodiments, the present invention relates to the treatment of anemia is a patient having an acute kidney injury (AKI).
  • In various embodiments, the present compositions and methods are used to reduce or eliminate fatigue, dizziness, and shortness of breath in a patient.
  • In various embodiments, the present compositions and methods are used to treat a patient presenting with hyporesponse or resistance to erythropoiesis stimulating agent therapy. In some embodiments, hyporesponsiveness to erythropoietin or ESA-resistant anemia refers to the presence of at least one of the following conditions: i) a significant decrease in hemoglobin levels at a constant dose of ESA treatment, ii) a significant increase in the ESA dose requirement to achieve or maintain a certain hemoglobin level, iii) a failure to raise the hemoglobin level to the target range despite the ESA dose equivalent to erythropoietin greater than 150 IU/kg/week or 0.75 mg/kg/week of darbepoeitn-alpha or continued need for such high dose of ESA to maintain the target hemoglobin level. For example, approximately 5-10% of patients with CDK demonstrate hyporesponsiveness to ESA, defined as a continued need for greater than 300 IU/kg per week erythropoietin or 1.5 ng/kg per week darbepoetin administered by the subcutaneous route.
  • In various embodiments, the present compositions and methods mitigate the need for dose-escalation of the erythropoiesis stimulating agent therapy and therefore, optionally, avoid side effects (e.g. flu-like symptoms such as joint pains, weakness, dizziness and tiredness, skin irritation, increased risk of adverse cardiovascular complications).
  • In various embodiments, the present compositions and methods are used to maintain a hemoglobin level of about 12.5 to 13 g/dL. In various embodiments, the present compositions and methods are used in patients having hemoglobin levels of below about 12 g/dL, or about 11 g/dL, or about 10 g/dL, or about 9 g/dL, or about 8 g/dL, or about 7 g/dL, or about 6 g/dL, or about 5 g/dL. In various embodiments, the present compositions and methods are used in patients having iron blood test scores that indicate blood pathology, e.g. a ferritin score of below about 200 ng/L and/or a transferrin saturation score below about 30%.
  • In various embodiments, the present compositions and methods are used to increase or maintain hemoglobin levels at a target level ranging from 9 to 10 g/dL, at a target level ranging from 9 g/dL to 11 g/dL, at a target level ranging from 9 g/dL to 12 g/dL, at a target level ranging from 9 g/dL to 14 g/dL, at a target level ranging from 10 g/dL to 14 g/dL, or at a target level ranging from 12 g/dL to 14 g/dL.
  • In various embodiments, the present compositions and methods are used to bring a patient's hemoglobin levels to normal. In various embodiments, normal hemoglobin ranges for humans are about 14-18 g/dl for men and 12-16 for women g/dl with the average hemoglobin value for men at about 16 g/dL and for women at about 14 g/dL.
  • In some embodiments, for instance when targeting EPO, the present invention relates to the treatment of anemia of one or more of the following toxicity grading criteria (e.g. NCI Common Toxicity Criteria): grade 1 (mild), 10.0 g hemoglobin/dL to within normal limits; grade 2 (moderate), 8.0-10.0 g of hemoglobin/dL; grade 3 (serious or severe), 6.5-7.9 g of hemoglobin/dL; and grade 4 (life threatening), less than 6.5 g of hemoglobin/dL. In various embodiments, the present invention brings an increase in toxicity grading criteria by about 1 point, or about 2 points, or about 3 points, or about 4 points. In various embodiments, the present invention results in a patient having a level of 0 or 1. In various embodiments, the present compositions and methods improve anemia as assessed by one or more scales described in Groopman et al. J Natl Cancer Inst (1999) 91 (19): 1616-1634, the entire contents of which are hereby incorporated by reference in their entireties.
  • In some embodiments, when targeting EPO, the present invention relates to combination therapy with one or more EPOs. For example, the present compositions may provide a sustained effect that can supplement a fast action of another EPO. In some embodiments, the present compositions are used as an adjuvant to other EPOs. In some embodiments, the present compositions are used as a maintenance therapy to other EPOs Other EPOs include the following: epoetin alfa, including without limitation, DARBEPOETIN (ARANESP), EPOCEPT (LUPIN PHARMA), NANOKINE (NANOGEN PHARMACEUTICAL), EPOFIT (INTAS PHARMA), EPOGEN (AMGEN), EPOGIN, EPREX, (JANSSEN-CILAG), BINOCRIT (SANDOZ), PROCRIT; epoetin beta, including without limitation, NEORECORMON (HOFFMANN-LA ROCHE), RECORMON, Methoxy polyethylene glycol-epoetin beta (MIRCERA, ROCHE); epoetin delta, including without limitation, DYNEPO (erythropoiesis stimulating protein, SHIRE PLC); epoetin omega, including without limitation, EPOMAX; epoetin zeta, including without limitation, SILAPO (STADA) and RETACRIT (HOSPIRA) and other EPOs, including without limitation, EPOCEPT (LUPIN PHARMACEUTICALS), EPOTRUST (PANACEA BIOTEC LTD), ERYPRO SAFE (BIOCON LTD.), REPOITIN (SERUM INSTITUTE OF INDIA LIMITED), VINTOR (EMCURE PHARMACEUTICALS), EPOFIT (INTAS PHARMA), ERYKINE (INTAS BIOPHARMACEUTICA), WEPOX (WOCKHARDT BIOTECH), ESPOGEN (LG LIFE SCIENCES), RELIPOIETIN (RELIANCE LIFE SCIENCES), SHANPOIETIN (SHANTHA BIOTECHNICS LTD), ZYROP (CADILA HEALTHCARE LTD.), EPIAO (RHUEPO) (SHENYANG SUNSHINE PHARMACEUTICAL CO. LTD), CINNAPOIETIN (CINNAGEN).
  • In some embodiments, when targeting EPO, the present invention relates to combination therapy with a blood transfusion. For instance, the present compositions may supplement a blood transfusion. In some embodiments, when targeting EPO, the present invention relates to combination therapy with iron supplements.
  • In some embodiments, the present invention also relates to the following protein targets, e.g. in the treatment of disease: growth hormone (GH) e.g. human and bovine growth hormone, growth hormone-releasing hormones; interferon including α-, β-, or γ-interferons, etc., interleukin-I; interleukin-II; erythropoietin including α- and β-erythropoietin (EPO), granulocyte colony stimulating factor (GCSF), granulocyte macrophage colony stimulating factor (GM-CSF), anti-agiogenic proteins (e.g., angiostatin, endostatin) PACAP polypeptide (pituitary adenylate cyclase activating polypeptide), vasoactive intestinal peptide (VIP), thyrotrophin releasing hormone (TRH), corticotropin releasing hormone (CRH), vasopressin, arginine vasopressin (AVP), angiotensin, calcitonin, atrial naturetic factor, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, insulin, somatotropin, plasminogen tissue activator, coagulation factors including coagulation factors VIII and IX, glucosylceramidase, sargramostim, lenograstin, filgrastin, dornase-α, molgramostim, PEG-L-asparaginase, PEG-adenosine deaminase, hirudin, eptacog-α (human blood coagulation factor VIIa) nerve growth factors, transforming growth factor, epidermal growth factor, basic fibroblast growth factor, VEGF; heparin including low molecular weight heparin, calcitonin; antigens; monoclonal antibodies; vancomycin; desferrioxamine (DFO); parathyroid hormone, an immunogen or antigen, and an antibody such as a monoclonal antibody.
  • In some embodiments, the present methods allow for effective additional therapeutic agent (e.g. those described herein) activity and/or targeting to a cell and/or tissue of interest. For example, the present synthetic RNA can lead to increased expression of one or more targeting molecules that direct an additional therapeutic to the location of therapy. For example, the additional therapeutic agent may have a binding partner that the synthetic RNA encodes. For example, the synthetic RNA may induce the expression of an antigen that directs the therapeutic activity of an antibody that may be used in combination (e.g. herceptin, rituxan, campath, gemtuzumab, herceptin, panorex, rituximab, bexxar, edrecolomab, alemtuzumab, mylotrag, IMC-C225, smartin 195, and mitomomab). In some embodiments, the synthetic RNA can be injected directly into one or more of the tumors described herein and home the therapeutic antibody to the tumor.
  • In some embodiments, the present methods allow for effective additional therapeutic agent generation, especially when the additional therapeutic agent is a prodrug, for example, to produce an active form of the drug. In some embodiments, the synthetic RNA can be injected directly into one or more of the tumors described herein and home the prodrug to the tumor. For instance, the synthetic RNA may encode an enzyme that catalyzes the localized conversion of a non-toxic, systemically delivered agent into a potent chemotherapeutic agent. By way of illustration (note that any of the prodrugs or drugs listed herein are additional agents as used herein):
  • Enzyme Prodrug Drug
    aldehyde oxidases 5-Ethynyl-2(1H)-pyrimidinone 5-Ethynyluracil
    aldehyde oxidases IPdR IUdR
    aldehyde oxidases 5-FP 5-FU
    amino acid oxidases d-alanine Hydrogen peroxide
    amino acid oxidases SeCys conjugates Selenols and hydrogen peroxide
    cytochrome P450 reductase Menadione Semiquinone radical
    cytochrome P450 reductase Mitomycin C Quinone methide intermediate
    cytochrome P450 reductase Tirapazamine Nitroxide radical
    cytochrome P450 reductase EO9 Unidentified semiquinone radical
    DT-diaphorase Streptonigrin Unidentified
    DT-diaphorase Mitomycin C Quinone methide intermediate
    DT-diaphorase CB 1954 5-(Aziridin-1-yl)-4-hydroxyl-amino-2-
    nitrobenzamide
    DT-diaphorase Diaziquone Semiquinone radicala
    cytochrome P450 Ipomeanol Unidentified (possibly furan epoxide)
    cytochrome P450 Ftorafur (tegafur) 5-FU
    cytochrome P450 Dacarbazine HHMTIC
    cytochrome P450 Trofosfamide Trofosfamide mustard
    cytochrome P450 Ifosfamide Isophosphamide mustard
    cytochrome P450 Cyclophosphamide Phosphoramide mustard
    cytochrome P450 AQ4N AQ4
    tyrosinase 2,4-Dihydroxyphenylalanine 6-Hydroxydopa
    tyrosinase 4-S-CAP BQ
    tyrosinase GHB GBQ
    tyrosinase Substituted phenols Orthoquinones
    tyrosinase Phenyl mustards Phenol mustard
    tyrosinase Urea mustards Unidentified
    glutathione S-transferase TER286 Aziridinium agent
    glutathione S-transferase S-CPHC-ethylsulfoxide S-CPHC-glutathione
    glutathione S-transferase PTA 6-MP
    carboxylesterase CPT-11 SN-38
    carboxylesterase Paclitaxel-2-ethylcarbonate Paclitaxel
    carboxylesterase Capecitabine 5′-Deoxy-5-fluorocytidine (5-FU)
    carboxylesterase Tertiary amidomethyl esters Carboxylic acids and aminesa
    alkaline phosphatase Amifostine WR-1065
    alkaline phosphatase 3-AP phosphate 3-AP
    β-glucuronidase BHAMG pHAM
    β-glucuronidase Epirubicin-glucuronide Epirubicin
    β-glucuronidase HMR 1826 Doxorubicin
    β-glucuronidase DNR-GA3 Daunorubicin
    β-glucuronidase DOX-GA3 Doxorubicin
    β-glucuronidase Paclitaxel glucuronide Paclitaxel
    β-glucuronidase 5-FU glucuronide 5-FU
    cysteine conjugate β-lyase PC 6-MP
    cysteine conjugate β-lyase GC 6-Thioguanine
    cysteine conjugate β-lyase SeCys conjugate Selenol
    Nitroreductase CB 1954 5-(Aziridin-1-yl)-4-hydroxyl-amino-2-
    nitro-benzamide
    Cytochrome P450 4-Ipomeanol Unidentified (furan epoxide is
    speculated)
    Cytochrome P450 Ifosfamide Isophosphoramide mustard
    Cytochrome P450 Cyclophosphamide Phosphoramide mustard
    Purine-nucleoside Fludarabine 2-Fluoroadenine
    phosphorylase
    Purine-nucleoside MeP-dR MeP
    phosphorylase
    Thymidine kinase Ganciclovir Ganciclovir-triphosphate nucleotide
    Alkaline phosphatase Etoposide phosphate Etoposide
    Alkaline phosphatase Mitomycin C phosphate Mitomycin C
    Alkaline phosphatase POMP POM
    Alkaline phosphatase N-(4-phosphonooxy)- Doxorubicin
    phenylacetyl)doxorubicin
    β-Glucuronidase Glucuronidated Nornitrogen mustard Oxazolidinone
    β-Glucuronidase Glucuronidated 9-amino- 9-Aminocamptothecin
    camptothecin
    β-Glucuronidase Glucuronide mustard Mustard
    Carboxypeptidase Methotrexate-amino acids Methotrexate
    Carboxypeptidase CMDA Benzoic acid mustard
    Penicillin amidase DPO Doxorubicin
    Penicillin amidase MelPO Melphalan
    Penicillin amidase NHPAP Palytoxin
    Penicillin amidase N-(phenylacetyl) doxorubicin Doxorubicin
    Penicillin amidase N-(phenylacetyl) melphalan Melphalan
    β-Lactamase LY 266070 DAVLBHYD
    β-Lactamase C-DOX Doxorubicin
    β-Lactamase PRODOX Doxorubicin
    β-Lactamase CM Phenylenediamine mustard
    β-Lactamase CCM Phenylenediamine mustard
    β-Lactamase Cephalosporin-DACCP DACCP
    β-Lactamase PROTAX Taxol
    β-Lactamase Cephalosporin mitomycin C Mitomycin C
    β-Lactamase C-Mel Melphalan
    Cytosine deaminase 5-Fluorocytosine 5-Fluorouracil
    Methionine γ-lyase Selenomethionine Methylselenol
    Methionine γ-lyase Trifluoromethionine CSF2
  • In certain embodiments, the synthetic RNA may encode an enzyme that catalyzes the conversion of 5-FU and/or Doxorubicin from various prodrugs, as illustrated by the examples below:
  • 5-FU Prodrugs and Enzymes
    5-FP Aldehyde oxidase
    Ftorafur P450
    5′-DFUR Thymidine phosphorylase
    5-FU glucuronide β-Glucuronidase
    5-FC Cytosine deaminase
    Doxorubicin Prodrugs and Enzymes
    N-(4-phosphono-oxy)-phenylacetyl) Alkaline phosphatase
    doxorubicin
    HMR 1826 β-Glucuronidase
    DOX-GA3 β-Glucuronidase
    DPO Penicillin amidase
    N-(phenyacetyl) doxorubicin Penicillin amidase
    C-DOX β-Lactamase
    PRODOX β-Lactamase
  • In some embodiments, the nucleic acid drugs at the doses and regimens described herein may be used in combination with one or more additional agents (aka adjuvant therapy or combination agent). In some embodiments, the nucleic acid drugs at the doses and regimens described herein may be used in a human patient undergoing treatment with one or more additional agents. In some embodiments, the nucleic acid drug is used as an adjuvant or neoadjuvant to any of the additional agents described herein. In some embodiments, the invention pertains to co-administration and/or co-formulation. Any of the compositions described herein may be co-formulated and/or co-administered. In some embodiments, any nucleic acid drug described herein acts synergistically when co-administered with another agent and may be administered at doses that are lower than the doses commonly employed when such agents are used as monotherapy.
  • In some embodiments, any nucleic acid drug described herein may include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the composition such that covalent attachment does not prevent the activity of the composition. For example, but not by way of limitation, derivatives include composition that have been modified by, inter alia, glycosylation, lipidation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications can be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of turicamycin, etc. Additionally, the derivative can contain one or more non-classical amino acids. In various embodiments, one or more additional agents (aka adjuvant therapy or combination agent) may be conjugated to any nucleic acid drug described herein.
  • Contacting a cell with a steroid can suppress the innate immune response to foreign nucleic acids, and can increase the efficiency of nucleic acid delivery and translation. Certain embodiments are therefore directed to contacting a cell with a steroid. Other embodiments are directed to administering a steroid to a patient. Illustrative steroids include corticosteroid steriods. In some embodiments, the steroid is one or more of cortisone, hydrocortisone, prednisone, prednisolone, dexamethasone, triamcinolone, and betamethasone. In one embodiment, the steroid is hydrocortisone. In another embodiment, the steroid is dexamethasone.
  • Other embodiments are directed to administering to a patient a member of the group: an antibiotic, an antimycotic, and an RNAse inhibitor.
  • Botulinum toxin type A has been approved by the US Food and Drug Administration (FDA) for the treatment of essential blepharospasm, strabismus and hemifacial spasm in patients over the age of twelve, cervical dystonia, glabellar line (facial) wrinkles and for treating hyperhydrosis and botulinum toxin type B has been approved for the treatment of cervical dystonia. The present compositions may be combined with these toxins in the treatment of these diseases and related diseases. Some embodiments are directed to a nucleic acid drug targeting a neurotoxin. In various embodiments, the neurotoxin is a botulinum toxin or a biologically active fragment, variant, analogue or family-member thereof.
  • Further the combination of any one of the aforementioned toxins may be used in combination with the present compositions for various cosmetic procedures, including, without limitation, facial wrinkles, hyperkinetic skin lines, glabellar lines, crow's feet, marionette lines, skin disorders, nasolabial folds, blepharospasm, strabismus, hemifacial spasms and sweating disorders. Alternatively, the present compositions may be used to in these cosmetic procedures as a monotherapy.
  • Certain embodiments are directed to a combination therapy comprising one or more of the therapeutic or cosmetic compositions of the present invention and one or more adjuvant therapies or cosmetic treatments. In certain embodiments, one or more of the therapeutic or cosmetic compositions of the present invention are administered to a subject which is undergoing treatment with one or more adjuvant therapies or cosmetic treatments. Example adjuvant therapies and cosmetic treatments are set forth in Table 3 and Table 5 of U.S. Provisional Application No. 61/721,302, the contents of which are hereby incorporated by reference, and are given by way of example, and not by way of limitation.
  • TABLE 3
    Illustrative Adjuvant Therapies
    Example
    Therapy/Treatment Class Disease/Condition Therapy/Treatment
    Acetylcholinesterase inhibitors Myasthenia gravis, Glaucoma, Edrophonium
    Alzheimer's disease, Lewy body
    dementia, Postural tachycardia syndrome
    Angiotensin-converting-enzyme Hypertension, Congestive heart failure Perindopril
    inhibitor
    Alkylating agents Cancer Cisplatin
    Angiogenesis inhibitors Cancer, Macular degeneration Bevacizumab
    Angiotensin II receptor antagonists Hypertension, Diabetic nephropathy, Valsartan
    Congestive heart failure
    Antibiotics Bacterial infection Amoxicillin
    Antidiabetic drugs Diabetes Metformin
    Antimetabolites Cancer, Infection 5-fluorouracil (5FU)
    Antisense oligonucleotides Cancer, Diabetes, Amyotrophic lateral Mipomersen
    sclerosis (ALS), Hypercholesterolemia
    Cytotoxic antibiotics Cancer Doxorubicin
    Deep-brain stimulation Chronic pain, Parkinson's disease, N/A
    Tremor, Dystonia
    Dopamine agonists Parkinson's disease, Type II diabetes, Bromocriptine
    Pituitary tumors
    Entry/Fusion inhibitors HIV/AIDS Maraviroc
    Glucagon-like peptide-1 agonists Diabetes Exenatide
    Glucocorticoids Asthma, Adrenal insufficiency, Dexamethasone
    Inflammatory diseases, Immune diseases,
    Bacterial meningitis
    Immunosuppressive drugs Organ transplantation, Inflammatory Azathioprine
    diseases, Immune diseases
    Insulin/Insulin analogs Diabetes NPH insulin
    Integrase inhibitors HIV/AIDS Raltegravir
    MAO-B inhibitors Parkinson's disease, Depression, Selegiline
    Dementia
    Maturation inhibitors HIV/AIDS Bevirimat
    Nucleoside analog reverse- HIV/AIDS, Hepatitis B Lamivudine
    transcriptase inhibitors
    Nucleotide analog reverse- HIV/AIDS, Hepatitis B Tenofovir
    transcriptase inhibitors
    Non-nucleoside reverse- HIV/AIDS Rilpivirine
    transcriptase inhibitors
    Pegylated interferon Hepatitis B/C, Multiple sclerosis Interferon beta-1a
    Plant alkaloids/terpenoids Cancer Paclitaxel
    Protease inhibitors HIV/AIDS, Hepatitis C, Other viral Telaprevir
    infections
    Radiotherapy Cancer Brachytherapy
    Renin inhibitors Hypertension Aliskiren
    Statins Hypercholesterolemia Atorvastatin
    Topoisomerase inhibitors Cancer Topotecan
    Vasopressin receptor antagonist Hyponatremia, Kidney disease Tolvaptan
    Dermal filler Wrinkles, aged skin Hyaluronic Acid
    Botulinum toxin Wrinkles, aged skin botulinum toxin type A
    Induction of skin repair Acne scars, aged skin Laser treatment,
    dermabrasion
  • In some embodiments, the additional agent is a cytotoxic agent, comprising, in illustrative embodiments, a toxin, a chemotherapeutic agent, a radioisotope, and an agent that causes apoptosis or cell death.
  • Illustrative cytotoxic agents include, but are not limited to, methotrexate, aminopterin, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine; alkylating agents such as mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU), mitomycin C, lomustine (CCNU), 1-methylnitrosourea, cyclothosphamide, mechlorethamine, busulfan, dibromomannitol, streptozotocin, mitomycin C, cis-dichlorodiamine platinum (II) (DDP) cisplatin and carboplatin (paraplatin); anthracyclines include daunorubicin (formerly daunomycin), doxorubicin (adriamycin), detorubicin, carminomycin, idarubicin, epirubicin, mitoxantrone and bisantrene; antibiotics include dactinomycin (actinomycin D), bleomycin, calicheamicin, mithramycin, and anthramycin (AMC); and antimytotic agents such as the vinca alkaloids, vincristine and vinblastine. Other cytotoxic agents include paclitaxel (taxol), ricin, pseudomonas exotoxin, gemcitabine, cytochalasin B, gramicidin D, ethidium bromide, emetine, etoposide, tenoposide, colchicin, dihydroxy anthracin dione, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, procarbazine, hydroxyurea, asparaginase, corticosteroids, mytotane (O,P′-(DDD)), interferons, and mixtures of these cytotoxic agents.
  • Further cytotoxic agents include, but are not limited to, chemotherapeutic agents such as carboplatin, cisplatin, paclitaxel, gemcitabine, calicheamicin, doxorubicin, 5-fluorouracil, mitomycin C, actinomycin D, cyclophosphamide, vincristine, bleomycin, VEGF antagonists, EGFR antagonists, platins, taxols, irinotecan, 5-fluorouracil, gemcytabine, leucovorine, steroids, cyclophosphamide, melphalan, vinca alkaloids (e.g., vinblastine, vincristine, vindesine and vinorelbine), mustines, tyrosine kinase inhibitors, radiotherapy, sex hormone antagonists, selective androgen receptor modulators, selective estrogen receptor modulators, PDGF antagonists, TNF antagonists, IL-1 antagonists, interleukins (e.g. IL-12 or IL-2), IL-12R antagonists, Toxin conjugated monoclonal antibodies, tumor antigen specific monoclonal antibodies, Erbitux, Avastin, Pertuzumab, anti-CD20 antibodies, Rituxan, ocrelizumab, ofatumumab, DXL625, HERCEPTIN, or any combination thereof. Toxic enzymes from plants and bacteria such as ricin, diphtheria toxin and Pseudomonas toxin may be conjugated to the therapeutic agents (e.g. antibodies) to generate cell-type-specific-killing reagents (Youle, et al., Proc. Nat'l Acad. Sci. USA 77:5483 (1980); Gilliland, et al., Proc. Nat'l Acad. Sci. USA 77:4539 (1980); Krolick, et al., Proc. Nat'l Acad. Sci. USA 77:5419 (1980)).
  • Other cytotoxic agents include cytotoxic ribonucleases as described by Goldenberg in U.S. Pat. No. 6,653,104. Embodiments of the invention also relate to radioimmunoconjugates where a radionuclide that emits alpha or beta particles is stably coupled to the antibody, or binding fragments thereof, with or without the use of a complex-forming agent. Such radionuclides include beta-emitters such as Phosphorus-32, Scandium-47, Copper-67, Gallium-67, Yttrium-88, Yttrium-90, Iodine-125, Iodine-131, Samarium-153, Lutetium-177, Rhenium-186 or Rhenium-188, and alpha-emitters such as Astatine-211, Lead-212, Bismuth-212, Bismuth-213 or Actinium-225.
  • Illustrative detectable moieties further include, but are not limited to, horseradish peroxidase, acetylcholinesterase, alkaline phosphatase, beta-galactosidase and luciferase. Further exemplary fluorescent materials include, but are not limited to, rhodamine, fluorescein, fluorescein isothiocyanate, umbelliferone, dichlorotriazinylamine, phycoerythrin and dansyl chloride. Further exemplary chemiluminescent moieties include, but are not limited to, luminol. Further exemplary bioluminescent materials include, but are not limited to, luciferin and aequorin. Further exemplary radioactive materials include, but are not limited to, Iodine-125, Carbon-14, Sulfur-35, Tritium and Phosphorus-32.
  • The dosage of any additional agent described herein as well as the dosing schedule can depend on various parameters, including, but not limited to, the human patient's general health, and the administering physician's and/or human patient's discretion. Co-administration may be simultaneous or sequential. Any additional agent described herein, can be administered prior to (e.g., about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 12 hours, about 24 hours, about 48 hours, about 72 hours, about 96 hours, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, 8 weeks, or about 12 weeks before), concurrently with, or subsequent to (e.g., about 5 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 4 hours, about 6 hours, about 12 hours, about 24 hours, about 48 hours, about 72 hours, about 96 hours, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 8 weeks, or about 12 weeks after) the administration of the nucleic acid drug to a human patient in need thereof. In various embodiments any agent described herein is administered about 1 minute apart, about 10 minutes apart, about 30 minutes apart, less than about 1 hour apart, about 1 hour apart, about 1 hour to about 2 hours apart, about 2 hours to about 3 hours apart, about 3 hours to about 4 hours apart, about 4 hours to about 5 hours apart, about 5 hours to about 6 hours apart, about 6 hours to about 7 hours apart, about 7 hours to about 8 hours apart, about 8 hours to about 9 hours apart, about 9 hours to about 10 hours apart, about 10 hours to about 11 hours apart, about 11 hours to about 12 hours apart, no more than about 24 hours apart or no more than about 48 hours apart.
  • In a specific embodiment, the combination regimen is designed to exploit the finding that the nucleic acid drug dosing and formulation of the present invention has potent effects quickly (e.g. in about 6, or about 12, or about 24, or about 30, or about 36, or about 48 hours) and the effects can last about 7 days, or longer.
  • The dose of a nucleic acid drug is disclosed herein. In general, the dose of any additional agent that is useful is known to those in the art. For example, doses may be determined with reference Physicians' Desk Reference, 66th Edition, PDR Network; 2012 Edition (Dec. 27, 2011), the contents of which are incorporated by reference in its entirety. In some embodiments, the present invention allows a patient to receive doses that exceed those determined with reference Physicians' Desk Reference. The dosage of any additional agent described herein can depend on several factors including the severity of the condition, whether the condition is to be treated or prevented, and the age, weight, and health of the human patient to be treated. Additionally, pharmacogenomic (the effect of genotype on the pharmacokinetic, pharmacodynamic or efficacy profile of a therapeutic) information about a particular human patient may affect dosage used. Furthermore, the exact individual dosages can be adjusted somewhat depending on a variety of factors, including the specific combination of the agents being administered, the time of administration, the route of administration, the nature of the formulation, the rate of excretion, the particular disease being treated, the severity of the disorder, and the anatomical location of the disorder. Some variations in the dosage can be expected.
  • Cells, tissues, organs, and organisms, including, but not limited to, humans, have several characteristics that can inhibit or prevent the delivery of nucleic acids, including, for example, the stratum corneum, which can serve as a barrier to foreign organisms and nucleic acids. These characteristics can thus inhibit the effects of therapeutics and cosmetics comprising nucleic acids. It has now been discovered that many of these characteristics can be circumvented or overcome using a patch comprising a flexible membrane and a plurality of needles, and that such a patch can serve as an effective and safe article for the delivery of nucleic acids. Certain embodiments are therefore directed to a nucleic acid delivery patch. In one embodiment, the nucleic acid delivery patch comprises a flexible membrane. In another embodiment, the nucleic acid delivery patch comprises a plurality of needles. In yet another embodiment, the plurality of needles are attached to the flexible membrane. In some embodiments, the patch comprises a nucleic acid. In one embodiment, the nucleic acid is present in solution. In one embodiment, the plurality of needles include one or more needles having a lumen. In another embodiment, the patch further comprises a second flexible membrane. In yet another embodiment, the flexible membrane and the second flexible membrane are arranged to form a cavity. In a further embodiment, the cavity contains a nucleic acid. In a still further embodiment, the membrane comprises one or more holes through which a nucleic acid can pass. In a still further embodiment, one or more holes and one or more needles having a lumen are arranged to allow the passage of a solution containing a nucleic acid through at least one of the one or more holes and through at least one of the one or more needles having a lumen. In some embodiments, the patch is configured to deliver a solution to the skin. In one embodiment, the solution comprises a nucleic acid. In another embodiment, the solution comprises a vehicle. In yet another embodiment, the vehicle is a lipid or lipidoid. In a still further embodiment, the vehicle is a lipid-based transfection reagent.
  • The cell membrane can serve as a barrier to foreign nucleic acids. It has now been discovered that combining the patch of the present invention with an electric field can increase the efficiency of nucleic acid delivery. Certain embodiments are therefore directed to a nucleic acid delivery patch comprising a plurality of needles, wherein at least two needles form part of a high-voltage circuit. Certain embodiments are directed to an implantable “tattoo” for microneedle delivery (see, e.g. Nature Materials 12, pp 367-376 (2013), the contents of which are hereby incorporated by reference in their entirety). In one embodiment, the high-voltage circuit generates a voltage greater than about 10V. In another embodiment, the high-voltage circuit generates a voltage greater than about 20V. In yet another embodiment, an electric field is produced between two of the needles. In a further embodiment, the magnitude of the electric field is at least about 100 V/cm. In a still further embodiment, the magnitude of the electric field is at least about 200 V/cm. In some embodiments, the patch is configured to deliver a nucleic acid to the epidermis. In other embodiments, the patch is configured to deliver a nucleic acid to the dermis. In still other embodiments, the patch is configured to deliver a nucleic acid to sub-dermal tissue. In still other embodiments, the patch is configured to deliver a nucleic acid to muscle. Certain embodiments are directed to a nucleic acid delivery patch comprising a plurality of electrodes. In one embodiment, the plurality of electrodes is attached to a flexible membrane. Other embodiments are directed to a nucleic acid delivery patch comprising a rigid structure. In one embodiment, a plurality of electrodes are attached to the rigid structure.
  • In some embodiments, the compositions described herein are administered using an array of needles covering an affected area of the subject. In some embodiments, the treatment area is mechanically massaged after administration. In some embodiments, the treatment area is exposed to electric pulses after administration. In some embodiments, the electric pulses are between about 10V and about 200V for from about 50 microseconds to about 1 second. In some embodiments, the electric pulses are generated around the treatment area by a multielectrode array.
  • In some embodiments, the present invention provides a patch delivery system, comprising a non-viral RNA transfection composition enclosed within a membrane, and an array of delivery needles delivering from about 10 ng to about 2000 ng of RNA per treatment area of about 100 cm2 or less, or about 50 cm2 or less, or about 10 cm2 or less, or about 5 cm2 or less, or about 1 cm2 or less, or about 0.5 cm2 or less, or about 0.2 cm2 or less. In some embodiments, the non-viral transfection composition contains from about 10 ng to about 2000 ng per injection volume of about 20 μL to about 1 ml. In some embodiments, each needle delivers an injection volume of between 1 μL and 500 μL.
  • In some embodiments, the delivery patch comprises an acrylic reservoir that holds the nucleic acid drug. In some embodiments, a silicon adhesive is added to create a semisolid suspension of microscopic, concentrated drug cells. Further, some embodiments pride a patch that is associated with one or more enhancers (these include, without limitation, iontophoresis, ultrasound, chemicals including gels, microneedles, sonophoresis, lasers, and electroporatic methods).
  • In some embodiments, the delivery is effected via a gel, optionally a hydro alcoholic gel containing a combination of enhancers (e.g. COMBIGEL (ANTARES PHARMA)).
  • In various embodiments, the RNA is delivered using needle arrays. Illustrative needle arrays include, but are not limited to AdminPen 600 and those described in U.S. Pat. Nos. 7,658,728, 7,785,301, and 8,414,548, the entire disclosure of which are hereby incorporated by reference. Other examples of needles include, for example, the 3M™ Hollow Microstructured Transdermal System and the 3M Solid Microstructured Transdermal Systems (sMTS). See, e.g. U.S. Pat. Nos. 3,034,507 and 3,675,766; Microneedles for Transdermal Drug Delivery. Advanced Drug Delivery Reviews. 56: 581-587 (2004); Pharm Res. 2011 Jan; 28(1): 31-40, the entire contents of which are hereby incorporated by reference in their entireties.
  • In some embodiments, microneedles and/or microneedle arrays may be used. In various embodiments, the microneedles and/or microneedle arrays may be, without limitation, solid, RNA-coated, dissolving, biodegradable, and/or hollow. In some embodiments, the delivery is effected via a microneedle system, optionally combined with an electronically controlled micropump that delivers the drug at specific times or upon demand. For example, the MACROFLUX (Alza) system may be used.
  • Other embodiments are directed to a method for delivering a nucleic acid to a cell in vivo comprising applying a nucleic acid to a tissue containing a cell in vivo. In one embodiment, the method further comprises applying a transient electric field in the vicinity of the cell. In another embodiment, the method results in the cell in vivo internalizing the nucleic acid. In yet another embodiment, the nucleic acid comprises synthetic RNA. In a further embodiment, the method further results in the cell internalizing a therapeutically or cosmetically effective amount of the nucleic acid. In one embodiment, the cell is a skin cell. In another embodiment, the cell is a muscle cell. In yet another embodiment, the cell is a dermal fibroblast. In a further embodiment, the cell is a keratinocyte. In a still further embodiment, the cell is a myoblast. In some embodiments, the nucleic acid comprises a protein of interest. In one embodiment, the protein of interest is a fluorescent protein. In another embodiment, the protein of interest is an extracellular-matrix protein. In yet another embodiment, the protein of interest is a member of the group: elastin, collagen, laminin, fibronectin, vitronectin, lysyl oxidase, elastin binding protein, a growth factor, fibroblast growth factor, transforming growth factor beta, granulocyte colony-stimulating factor, a matrix metalloproteinase, an actin, fibrillin, microfibril-associated glycoprotein, a lysyl-oxidase-like protein, platelet-derived growth factor, a lipase, an uncoupling protein, thermogenin, filaggrin, a fibroblast growth factor, an antibody, and a protein involved with pigment production. In some embodiments, the method further comprises delivering the nucleic acid to the epidermis. In other embodiments, the method further comprises delivering the nucleic acid to the dermis. In still other embodiments, the method further comprises delivering the nucleic acid below the dermis. In one embodiment, the delivering is by injection. In another embodiment, the delivering is by injection using a micro-needle array. In yet another embodiment, the delivering is by topical administration. In a further embodiment, the delivering comprises disruption or removal of a part of the tissue. In a still further embodiment, the delivering comprises disruption or removal of the stratum corneum. In some embodiments, the nucleic acid is present in solution. In one embodiment, the solution comprises a growth factor. In another embodiment, the growth factor is a member of the group: a fibroblast growth factor and a transforming growth factor. In yet another embodiment, the growth factor is a member of the group: basis fibroblast growth factor and transforming growth factor beta. In other embodiments, the solution comprises cholesterol.
  • In another embodiment, the method further comprises contacting the cell with one or more nucleic acid molecules. In yet another embodiment, at least one of the one or more nucleic acid molecules encodes a protein of interest. In a further embodiment, the method results in the cell expressing the protein of interest. In a still further embodiment, the method results in the cell expressing a therapeutically or cosmetically effective amount of the protein of interest.
  • In another embodiment, the cell is contacted with a nucleic acid molecule. In yet another embodiment, the method results in the cell internalizing the nucleic acid molecule. In a further embodiment, the method results in the cell internalizing a therapeutically or cosmetically effective amount of the nucleic acid molecule. In one embodiment, the nucleic acid encodes a protein of interest. In one embodiment, the nucleic acid molecule comprises a member of the group: a dsDNA molecule, a ssDNA molecule, a RNA molecule, a dsRNA molecule, a ssRNA molecule, a plasmid, an oligonucleotide, a synthetic RNA molecule, a miRNA molecule, an mRNA molecule, and an siRNA molecule. In various embodiments, the RNA comprises one or more non-canonical nucleotides.
  • In some embodiments, the present invention relates to one or more administration techniques described in U.S. Pat. Nos. 5,711,964; 5,891,468; 6,316,260; 6,413,544; 6,770,291; and 7,390,780, the entire contents of which are hereby incorporated by reference in their entireties.
  • The invention also provides kits that can simplify the administration of the nucleic acid drugs described herein and/or any additional agent described herein. An illustrative kit of the invention comprises a nucleic acid drug and/or any additional agent described herein in unit dosage form. In one embodiment, the unit dosage form is a container, such as a pre-filled syringe, which can be sterile, containing any agent described herein and a pharmaceutically acceptable carrier, diluent, excipient, or vehicle. The kit can further comprise a label or printed instructions instructing the use of any agent described herein. The kit or one or more components of the kit may be stored at room temperature, about 4° C., about −20° C., about −80° C., or about −196° C. The kit may also include a lid speculum, topical anesthetic, and a cleaning agent for the administration location. The kit can also further comprise one or more additional agent described herein. In one embodiment, the kit comprises a container containing an effective amount of a nucleic acid drug as disclosed herein and an effective amount of another composition, such as an additional agent as described herein. In some embodiments, the unit dosage form is a pre-loaded (a.k.a. pre-dosed or pre-filled) syringe or a pen needle injector (injection pen)). Such unit dosage forms may comprise the effective doses of nucleic acid drug described herein, e.g. about 10 ng to about 2000 ng, e.g. about 10 ng, or about 20 ng, or about 50 ng, or about 100 ng, or about 200 ng, or about 300 ng, or about 400 ng, or about 500 ng, or about 600 ng, or about 700 ng, or about 800 ng, or about 900 ng, or about 1000 ng, or about 1100 ng, or about 1200 ng, or about 1300 ng, or about 1400 ng, or about 1500 ng, or about 1600 ng, or about 1700 ng, or about 1800 ng, or about 1900 ng, or about 2000 ng.
  • Some embodiments are directed to synthetic RNA molecules with low toxicity and high translation efficiency. Other embodiments are directed to a cell-culture medium for high-efficiency in vivo transfection, reprogramming, and gene editing of cells. Other embodiments pertain to methods for producing synthetic RNA molecules encoding reprogramming proteins. Still further embodiments pertain to methods for producing synthetic RNA molecules encoding gene-editing proteins.
  • Some embodiments are directed to methods of gene-editing and/or gene correction. Some embodiments encompass synthetic RNA-based gene-editing and/or gene correction, e.g. with RNA comprising non-canonical nucleotides, e.g. RNA encoding one or more of a nuclease, a transcription activator-like effector nuclease (TALEN), a zinc-finger nuclease, a meganuclease, a nickase, a clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof. In some embodiments, the efficiency of the gene-editing and/or gene correction is high, for example, higher than DNA-based gene editing and/or gene correction. In some embodiments, the present methods of gene-editing and/or gene correction are efficient enough for in vivo application. In some embodiments, the present methods of gene-editing and/or gene correction are efficient enough to not require cellular selection (e.g. selection of cells that have been edited). In various embodiments, the efficiency of gene-editing of the present methods is about 1%, or about 2%, or about 3%, or about 4%, or about 5%, or about 6%, or about 7%, or about 8%, or about 9%, or about 10%, or about 20%, or about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 100%. In various embodiments, the efficiency of gene-correction of the present methods is about 1%, or about 2%, or about 3%, or about 4%, or about 5%, or about 6%, or about 7%, or about 8%, or about 9%, or about 10%, or about 20%, or about 30%, or about 840%, or about 950%, or about 60%, or about 70%, or about 80%, or about 90%, or about 100% Some embodiments are directed to high-efficiency gene-editing proteins comprising engineered nuclease cleavage domains. Other embodiments are directed to high-fidelity gene-editing proteins comprising engineered nuclease cleavage domains. Various embodiments are directed to high-efficiency gene-editing proteins comprising engineered DNA-binding domains. Other embodiments are directed to high-fidelity gene-editing proteins comprising engineered DNA-binding domains. Still other embodiments are directed to gene-editing proteins comprising engineered repeat sequences. Some embodiments are directed to gene-editing proteins comprising one or more CRISPR associated family members. Some embodiments are directed to methods for altering the DNA sequence of a cell by transfecting the cell with or inducing the cell to express a gene-editing protein. Other embodiments are directed to methods for altering the DNA sequence of a cell that is present in an in vitro culture. Still further embodiments are directed to methods for altering the DNA sequence of a cell that is present in vivo.
  • Some embodiments are directed to methods of modulating the secretion or subcellular localization of a polypeptide. In such embodiments, the present invention provides an RNA encoding a protein comprising a signal peptide operably linked to a polypeptide having biological functionality. In an embodiment, the signal peptide modulates the secretion of the polypeptide. In another embodiment, the signal peptide modulates the subcellular localization of the polypeptide. For example, BMP7 comprising at least one FGF21 signal peptide may result in increased secretion of BMP7. In another example, BMP7 comprising at least one FGF21 signal peptide in place of the endogenous BMP7 signal peptide may result in increased secretion of BMP7. In various embodiments, any of the proteins listed in Table 2A or Table 2B comprising at least one signal peptide listed in the table below may result in increased secretion of the proteins.
  • Protein Signal Peptide
    Gaussia luciferase MGVKVLFALICIAVAEA
    Human BMP7 MHVRSLRAAAPHSFVALWAPLFLLRSALA
    Human chymo- MAFLWLLSCWALLGTTFG
    trypsinogen B
    Human chymo- MLGITVLAALLACASS
    trypsinogen C
    Human EPO MGVHECPAWLWLLLSLLSLPLGLPVLG
    Human FGF19 MRSGCVVVHVWILAGLWLAVAGRP
    Human FGF21 MDSDETGFEHSGLWVSVLAGLLLGACQA
    Human FGF23 MLGARLRLWVCALCSVCSMSVLRA
    Human IL2 MYRMQLLSCIALSLALVTNS
    Human IL22 MAALQKSVSSFLMGTLATSCLLLLALLVQ
    GGAA
    Human IL6 MNSFSTSAFGPVAFSLGLLLVLPAAFPAP
    Human Interferon MALTFALLVALLVLSCKSSCSVG
    Alpha-2
    Human Interferon MTNKCLLQIALLLCFSTTALS
    Beta
    Human Interferon MKYTSYILAFQLCIVLGSLGCYC
    Gamma
    Human Trypsin-1 MNPLLILTFVAAALA
  • Some embodiments are directed to methods of modulating the serum half-life, secretion, bioavailability, and/or activity of a protein comprising administering a RNA encoding the protein and a RNA encoding a receptor for the protein. For example, IL15 may be administered with its receptor, e.g., IL15RA to enhance one or more of the serum half-life, secretion, bioavailability, and activity of IL15.
  • Many proteins and peptides, when translated from in vitro transcribed RNA, can exhibit reduced activity resulting from incomplete or inadequate post-translational processing. It has now been discovered that the amount of active protein, polypeptide or peptide produced following RNA transfection can be increased by contacting cells with and/or inducing cells to express a second protein that is capable of processing a polypeptide into the protein, peptide or polypeptide. Certain embodiments are therefore directed to a method for inducing a cell to express an active protein, polypeptide or peptide comprising contacting the cell with a synthetic RNA molecule encoding a first polypeptide and contacting the cell with a second protein that is capable of processing the first polypeptide into an active protein, polypeptide or peptide.
  • In certain embodiments, the second protein is administered to a patient. In one embodiment, the second protein is a recombinant protein. In another embodiment, the cells are contacted with a synthetic RNA molecule encoding the second protein. In a further embodiment, the cells are contacted with and/or induced to express an inhibitor of a molecule that inhibits the second protein. In one embodiment, the inhibitor is a short interfering RNA molecule.
  • In certain embodiments, the second protein is a member of the PCSK family. In other embodiments, the second protein is a proprotein convertase. In still other embodiments, the second protein is a prohormone convertase. In still other embodiments, the second protein is a carboxypeptidase. In one embodiment, the second protein is PCSK3 (Furin/PACE). In another embodiment, the second protein is primarily secreted. In yet another embodiment, the second protein is primarily intracellular. In a further embodiment, the first polypeptide, protein, polypeptide or peptide is selected from the group: a product of proopiomelanocortin, renin, a product of enkephalin, a product of prodynorphin, somatostatin, insulin, agouti-related peptide, glucagon, parathyroid hormone, a member of the transforming growth factor beta superfamily, albumin, beta-secretase 1, nerve growth factor, caldesmon, the alpha-integrins, factor IX, α-melanocyte-stimulating hormone, adrenocorticotropic hormone, β-endorphin, and met-enkefalin.
  • Glycation and glycosylation are processes by which one or more sugar molecules are bound to a protein. It has now been discovered that altering the number or location of glycation and glycosylation sites can increase or decrease the stability of a protein. Certain embodiments are therefore directed to a protein with one or more glycation or glycosylation sites. In one embodiment, the protein is engineered to have more glycation or glycosylation sites than a natural variant of the protein. In another embodiment, the protein is engineered to have fewer glycation or glycosylation sites than a natural variant of the protein. In yet another embodiment, the protein has increased stability. In yet another embodiment, the protein has decreased stability. In some embodiments, the protein is a circulating protein. In one embodiment, the protein is erythropoietin or a biologically active fragment, variant, analogue, or family-member thereof. In another embodiment, the protein is darbepoetin alfa or a biologically active fragment, variant, analogue, or family-member thereof. In another embodiment, the protein is NOVEPOETIN or a biologically active fragment, variant, analogue, or family-member thereof.
  • It has been further discovered that in certain situations, including one or more steroids and/or one or more antioxidants in the transfection medium can increase in vivo transfection efficiency, in vivo reprogramming efficiency, and in vivo gene-editing efficiency. Certain embodiments are therefore directed to contacting a cell or patient with a glucocorticoid, such as hydrocortisone, prednisone, prednisolone, methylprednisolone, dexamethasone or betamethasone. Other embodiments are directed to a method for inducing a cell to express a protein of interest by contacting a cell with a medium containing a steroid and contacting the cell with one or more nucleic acid molecules. In one embodiment, the nucleic acid molecule comprises synthetic RNA. In another embodiment, the steroid is hydrocortisone. In yet another embodiment, the hydrocortisone is present in the medium at a concentration of between about 0.1 uM and about 10 uM, or about 1 uM. Other embodiments are directed to a method for inducing a cell in vivo to express a protein of interest by contacting the cell with a medium containing an antioxidant and contacting the cell with one or more nucleic acid molecules. In one embodiment, the antioxidant is ascorbic acid or ascorbic-acid-2-phosphate. In another embodiment, the ascorbic acid or ascorbic-acid-2-phosphate is present in the medium at a concentration of between about 0.5 mg/L and about 500 mg/L, including about 50 mg/L. Still other embodiments are directed to a method for reprogramming and/or gene-editing a cell in vivo by contacting the cell with a medium containing a steroid and/or an antioxidant and contacting the cell with one or more nucleic acid molecules, wherein the one or more nucleic acid molecules encodes one or more reprogramming and/or gene-editing proteins. In certain embodiments, the cell is present in an organism, and the steroid and/or antioxidant are delivered to the organism.
  • Adding transferrin to the complexation medium has been reported to increase the efficiency of plasmid transfection in certain situations. It has now been discovered that adding transferrin to the complexation medium can also increase the efficiency of in vivo transfection with synthetic RNA molecules. Certain embodiments are therefore directed to a method for inducing a cell in vivo to express a protein of interest by adding one or more synthetic RNA molecules and a transfection reagent to a solution containing transferrin. In one embodiment, the transferrin is present in the solution at a concentration of between about 1 mg/L and about 100 mg/L, such as about 5 mg/L. In another embodiment, the transferrin is recombinant.
  • In certain situations, including pertaining to culturing, it may be desirable to replace animal-derived components with non-animal-derived and/or recombinant components, in part because non-animal-derived and/or recombinant components can be produced with a higher degree of consistency than animal-derived components, and in part because non-animal-derived and/or recombinant components carry less risk of contamination with toxic and/or pathogenic substances than do animal-derived components. Certain embodiments are therefore directed to a protein that is non-animal-derived and/or recombinant. Other embodiments are directed to a medium, wherein some or all of the components of the medium are non-animal-derived and/or recombinant.
  • Other embodiments are directed to a method for transfecting a cell in vivo. In one embodiment, a cell in vivo is transfected with one or more nucleic acids, and the transfection is performed using a transfection reagent, such as a lipid-based transfection reagent. In one embodiment, the one or more nucleic acids includes at least one RNA molecule. In another embodiment, the cell is transfected with one or more nucleic acids, and the one or more nucleic acids encodes at least one of: p53, TERT, an antibody, an extracellular matrix protein, a cytokine, a secreted protein, a membrane-bound protein, an enzyme, a gene-editing protein, a chromatin-modifying protein, a DNA-binding protein, a transcription factor, a histone deacetylase, a pathogen-associated molecular pattern, and a tumor-associated antigen or a biologically active fragment, analogue, variant or family-member thereof. In another embodiment, the cell is transfected repeatedly, such as at least about 2 times during about 10 consecutive days, or at least about 3 times during about 7 consecutive days, or at least about 4 times during about 6 consecutive days. Some embodiments are directed to a method for increasing expression of telomerase in one of a fibroblast, a hematopoietic stem cell, a mesenchymal stem cells, a cardiac stem cell, a hair follicle stem cell, a neural stem cell, an intestinal stem cell, an endothelial stem cell, an olfactory stem cell, a neural crest stem cell, a testicular cell, and a keratinocyte. Some embodiments are directed to a method for increasing the length of telomeres in one of a fibroblast, a hematopoietic stem cell, a mesenchymal stem cells, a cardiac stem cell, a hair follicle stem cell, a neural stem cell, an intestinal stem cell, an endothelial stem cell, an olfactory stem cell, a neural crest stem cell, a testicular cell, and a keratinocyte. Other embodiments are directed to a method for isolating a cell from a patient, contacting the cell with a nucleic acid drug encoding a component of telomerase (e.g., TERT), and reintroducing the cell to the patient. Various embodiments are directed to a method for increasing the replicative potential of a cell.
  • Reprogramming can be performed by transfecting cells with one or more nucleic acids encoding one or more reprogramming factors. Examples of reprogramming factors include, but are not limited to: Oct4 protein, Sox2 protein, Klf4 protein, c-Myc protein, I-Myc protein, TERT protein, Nanog protein, Lin28 protein, Utf1 protein, Aicda protein, miR200 micro-RNA, miR302 micro-RNA, miR367 micro-RNA, miR369 micro-RNA and biologically active fragments, analogues, variants and family-members thereof. Certain embodiments are therefore directed to a method for reprogramming a cell in vivo. In one embodiment, the cell in vivo is reprogrammed by transfecting the cell with one or more nucleic acids encoding one or more reprogramming factors. In one embodiment, the one or more nucleic acids includes an RNA molecule that encodes Oct4 protein. In another embodiment, the one or more nucleic acids also includes one or more RNA molecules that encodes Sox2 protein, Klf4 protein, and c-Myc protein. In yet another embodiment, the one or more nucleic acids also includes an RNA molecule that encodes Lin28 protein. In one embodiment, the cell is a human skin cell, and the human skin cell is reprogrammed to a pluripotent stem cell. In another embodiment, the cell is a human skin cell, and the human skin cell is reprogrammed to a glucose-responsive insulin-producing cell. Examples of other cells that can be reprogrammed and other cells to which a cell can be reprogrammed include, but are not limited to: skin cells, pluripotent stem cells, mesenchymal stem cells, β-cells, retinal pigmented epithelial cells, hematopoietic cells, cardiac cells, airway epithelial cells, neural stem cells, neurons, glial cells, bone cells, blood cells, and dental pulp stem cells. In one embodiment, the cell is contacted with a medium that supports the reprogrammed cell. In one embodiment, the medium also supports the cell.
  • Importantly, infecting skin cells with viruses encoding Oct4, Sox2, Klf4, and c-Myc, combined with culturing the cells in a medium that supports the growth of cardiomyocytes, has been reported to cause reprogramming of the skin cells to cardiomyocytes, without first reprogramming the skin cells to pluripotent stem cells (See Efs et al Nat Cell Biol. 2011; 13:215-22, the contents of which are hereby incorporated by reference). In certain situations, direct reprogramming (reprogramming one somatic cell to another somatic cell without first reprogramming the somatic cell to a pluripotent stem cell, also known as “transdifferentiation”) may be desirable, in part because culturing pluripotent stem cells can be time-consuming and expensive, the additional handling involved in establishing and characterizing a stable pluripotent stem cell line can carry an increased risk of contamination, and the additional time in culture associated with first producing pluripotent stem cells can carry an increased risk of genomic instability and the acquisition of mutations, including point mutations, copy-number variations, and karyotypic abnormalities. Certain embodiments are therefore directed to a method for reprogramming a somatic cell in vivo, wherein the cell is reprogrammed to a somatic cell, and wherein a characterized pluripotent stem-cell line is not produced.
  • It has been further discovered that, in certain situations, fewer total transfections may be required to reprogram a cell according to the methods of the present invention than according to other methods. Certain embodiments are therefore directed to a method for reprogramming a cell in vivo, wherein between about 1 and about 12 transfections are performed during about 20 consecutive days, or between about 4 and about 10 transfections are performed during about 15 consecutive days, or between about 4 and about 8 transfections are performed during about 10 consecutive days. It is recognized that when a cell is contacted with a medium containing nucleic acid molecules, the cell may likely come into contact with and/or internalize more than one nucleic acid molecule either simultaneously or at different times. A cell can therefore be contacted with a nucleic acid more than once, e.g. repeatedly, even when a cell is contacted only once with a medium containing nucleic acids.
  • Of note, nucleic acids can contain one or more non-canonical or “modified” residues as described herein. For instance, any of the non-canonical nucleotides described herein can be used in the present reprgramming methods. In one embodiment, pseudouridine-5′-triphosphate can be substituted for uridine-5′-triphosphate in an in vitro-transcription reaction to yield synthetic RNA, wherein up to 100% of the uridine residues of the synthetic RNA may be replaced with pseudouridine residues. In vitro-transcription can yield RNA with residual immunogenicity, even when pseudouridine and 5-methylcytidine are completely substituted for uridine and cytidine, respectively (see, e.g., Angel. Reprogramming Human Somatic Cells to Pluripotency Using RNA [Doctoral Thesis]. Cambridge, Mass.: MIT; 2011, the contents of which are hereby incorporated by reference). For this reason, it is common to add an immunosuppressant to the transfection medium when transfecting cells with RNA. In certain situations, adding an immunosuppressant to the transfection medium may not be desirable, in part because the recombinant immunosuppressant most commonly used for this purpose, B18R, can be expensive and difficult to manufacture. It has now been discovered that cells in vivo can be transfected and/or reprogrammed according to the methods of the present invention, without using B18R or any other immunosuppressant. It has been further discovered that reprogramming cells in vivo according to the methods of the present invention without using immunosuppressants can be rapid, efficient, and reliable. Certain embodiments are therefore directed to a method for transfecting a cell in vivo, wherein the transfection medium does not contain an immunosuppressant. Other embodiments are directed to a method for reprogramming a cell in vivo, wherein the transfection medium does not contain an immunosuppressant. In certain situations, for example when using a high cell density, it may be beneficial to add an immunosuppressant to the transfection medium. Certain embodiments are therefore directed to a method for transfecting a cell in vivo, wherein the transfection medium contains an immunosuppressant. Other embodiments are directed to a method for reprogramming a cell in vivo, wherein the transfection medium contains an immunosuppressant. In one embodiment, the immunosuppressant is B18R or a biologically active fragment, analogue, variant or family-member thereof or dexamethasone or a derivative thereof. In one embodiment, the transfection medium does not contain an immunosuppressant, and the nucleic-acid dose is chosen to prevent excessive toxicity. In another embodiment, the nucleic-acid dose is less than about 1 mg/cm2 of tissue or less than about 1 mg/100,000 cells or less than about 10 mg/kg.
  • Reprogrammed cells produced according to certain embodiments of the present invention are suitable for therapeutic and/or cosmetic applications as they do not contain undesirable exogenous DNA sequences, and they are not exposed to animal-derived or human-derived products, which may be undefined, and which may contain toxic and/or pathogenic contaminants. Furthermore, the high speed, efficiency, and reliability of certain embodiments of the present invention may reduce the risk of acquisition and accumulation of mutations and other chromosomal abnormalities. Certain embodiments of the present invention can thus be used to generate cells that have a safety profile adequate for use in therapeutic and/or cosmetic applications. For example, reprogramming cells using RNA and the medium of the present invention, wherein the medium does not contain animal or human-derived components, can yield cells that have not been exposed to allogeneic material. Certain embodiments are therefore directed to a reprogrammed cell that has a desirable safety profile. In one embodiment, the reprogrammed cell has a normal karyotype. In another embodiment, the reprogrammed cell has fewer than about 5 copy-number variations (CNVs) relative to the patient genome, such as fewer than about 3 copy-number variations relative to the patient genome, or no copy-number variations relative to the patient genome. In yet another embodiment, the reprogrammed cell has a normal karyotype and fewer than about 100 single nucleotide variants in coding regions relative to the patient genome, or fewer than about 50 single nucleotide variants in coding regions relative to the patient genome, or fewer than about 10 single nucleotide variants in coding regions relative to the patient genome.
  • Endotoxins and nucleases can co-purify and/or become associated with other proteins, such as serum albumin. Recombinant proteins, in particular, can often have high levels of associated endotoxins and nucleases, due in part to the lysis of cells that can take place during their production. Endotoxins and nucleases can be reduced, removed, replaced or otherwise inactivated by many of the methods of the present invention, including, for example, by acetylation, by addition of a stabilizer such as sodium octanoate, followed by heat treatment, by the addition of nuclease inhibitors to the albumin solution and/or medium, by crystallization, by contacting with one or more ion-exchange resins, by contacting with charcoal, by preparative electrophoresis or by affinity chromatography. It has now been discovered that partially or completely reducing, removing, replacing or otherwise inactivating endotoxins and/or nucleases from a medium and/or from one or more components of a medium can increase the efficiency with which cells can be transfected and reprogrammed. Certain embodiments are therefore directed to a method for transfecting a cell in vivo with one or more nucleic acids, wherein the transfection medium is treated to partially or completely reduce, remove, replace or otherwise inactivate one or more endotoxins and/or nucleases. Other embodiments are directed to a medium that causes minimal degradation of nucleic acids. In one embodiment, the medium contains less than about 1 EU/mL, or less than about 0.1 EU/mL, or less than about 0.01 EU/mL.
  • In certain situations, protein-based lipid carriers such as serum albumin can be replaced with non-protein-based lipid carriers such as methyl-beta-cyclodextrin. The medium of the present invention can also be used without a lipid carrier, for example, when transfection is performed using a method that may not require or may not benefit from the presence of a lipid carrier, for example, using one or more lipid-based transfection reagents, polymer-based transfection reagents or peptide-based transfection reagents or using electroporation. Many protein-associated molecules, such as metals, can be highly toxic to cells in vivo. This toxicity can cause decreased viability, as well as the acquisition of mutations. Certain embodiments thus have the additional benefit of producing cells that are free from toxic molecules.
  • The associated-molecule component of a protein can be measured by suspending the protein in solution and measuring the conductivity of the solution. Certain embodiments are therefore directed to a medium that contains a protein, wherein about a 10% solution of the protein in water has a conductivity of less than about 500 μmho/cm. In one embodiment, the solution has a conductivity of less than about 50 μmho/cm. In another embodiment, less than about 0.65% of the dry weight of the protein comprises lipids and/or less than about 0.35% of the dry weight of the protein comprises free fatty acids.
  • The amount of nucleic acid delivered to cells in vivo can be increased to increase the desired effect of the nucleic acid. However, increasing the amount of nucleic acid delivered to cells in vivo beyond a certain point can cause a decrease in the viability of the cells, due in part to toxicity of the transfection reagent. It has now been discovered that when a nucleic acid is delivered to a population of cells in vivo in a fixed volume (for example, cells in a region of tissue), the amount of nucleic acid delivered to each cell can depend on the total amount of nucleic acid delivered to the population of cells and to the density of the cells, with a higher cell density resulting in less nucleic acid being delivered to each cell. In certain embodiments, a cell in vivo is transfected with one or more nucleic acids more than once. Under certain conditions, for example when the cells are proliferating, the cell density may change from one transfection to the next. Certain embodiments are therefore directed to a method for transfecting a cell in vivo with a nucleic acid, wherein the cell is transfected more than once, and wherein the amount of nucleic acid delivered to the cell is different for two of the transfections. In one embodiment, the cell proliferates between two of the transfections, and the amount of nucleic acid delivered to the cell is greater for the second of the two transfections than for the first of the two transfections. In another embodiment, the cell is transfected more than twice, and the amount of nucleic acid delivered to the cell is greater for the second of three transfections than for the first of the same three transfections, and the amount of nucleic acid delivered to the cells is greater for the third of the same three transfections than for the second of the same three transfections. In yet another embodiment, the cell is transfected more than once, and the maximum amount of nucleic acid delivered to the cell during each transfection is sufficiently low to yield at least about 80% viability for at least two consecutive transfections.
  • It has now been discovered that modulating the amount of nucleic acid delivered to a population of proliferating cells in vivo in a series of transfections can result in both an increased effect of the nucleic acid and increased viability of the cells. It has been further discovered that, in certain situations, when cells in vivo are contacted with one or more nucleic acids encoding one or more reprogramming factors in a series of transfections, the efficiency of reprogramming can be increased when the amount of nucleic acid delivered in later transfections is greater than the amount of nucleic acid delivered in earlier transfections, for at least part of the series of transfections. Certain embodiments are therefore directed to a method for reprogramming a cell in vivo, wherein one or more nucleic acids is repeatedly delivered to the cell in a series of transfections, and the amount of the nucleic acid delivered to the cell is greater for at least one later transfection than for at least one earlier transfection. In one embodiment, the cell is transfected between about 2 and about 10 times, or between about 3 and about 8 times, or between about 4 and about 6 times. In another embodiment, the one or more nucleic acids includes at least one RNA molecule, the cell is transfected between about 2 and about 10 times, and the amount of nucleic acid delivered to the cell in each transfection is the same as or greater than the amount of nucleic acid delivered to the cell in the most recent previous transfection. In yet another embodiment, the amount of nucleic acid delivered to the cell in the first transfection is between about 20 ng/cm2 and about 250 ng/cm2, or between 100 ng/cm2 and 600 ng/cm2. In yet another embodiment, the cell is transfected about 5 times at intervals of between about 12 and about 48 hours, and the amount of nucleic acid delivered to the cell is about 25 ng/cm2 for the first transfection, about 50 ng/cm2 for the second transfection, about 100 ng/cm2 for the third transfection, about 200 ng/cm2 for the fourth transfection, and about 400 ng/cm2 for the fifth transfection. In yet another embodiment, the cell is further transfected at least once after the fifth transfection, and the amount of nucleic acid delivered to the cell is about 400 ng/cm2.
  • Certain embodiments are directed to a method for transfecting a cell in vivo with a nucleic acid, wherein the amount of nucleic acid is determined by measuring the cell density, and choosing the amount of nucleic acid to transfect based on the measurement of cell density. In one embodiment, the cell density is measured by optical means. In another embodiment, the cell is transfected repeatedly, the cell density increases between two transfections, and the amount of nucleic acid transfected is greater for the second of the two transfections than for the first of the two transfections.
  • It has now been discovered that, in certain situations, the amount of a circulating protein that is produced in a patient can be increased by administering to a patient a nucleic acid at a plurality of administration sites. In certain embodiments, the amount of a circulating protein is increased relative to the amount of the circulating protein that is produced in a patient by administering to the patient the nucleic acid at a single injection site. In one embodiment, the administering is by injection. In another embodiment, the injection is intradermal injection. In still another embodiment, the injection is subcutaneous or intramuscular injection. In some embodiments, the plurality of administration sites comprise administration sites in the skin. In other embodiments, the plurality of administration sites are at least about 1 or at least about 2 or at least about 5 or at least about 10 or at least about 20 or at least about 50 or at least about 100 administration sites. In one embodiment, the administering is performed within at least about 5 minutes or at least about 10 minutes or at least about 30 minutes or at least about 1 hour or at least about 2 hours or at least about 5 hours or at least about 12 hours or at least about 1 day. In certain embodiments, the amount of a circulating protein is increased by at least about 10 percent or at least about 20 percent or at least about 50 percent or at least about 100 percent or at least about 3-fold or at least about 5-fold or at least about 10-fold or at least about 20-fold or at least about 50-fold or at least about 100-fold or at least about 500-fold or at least about 1000-fold or greater than 1000-fold.
  • It has now been discovered that, in certain situations, the in vivo transfection efficiency and viability of cells contacted with the medium of the present invention can be improved by conditioning the medium. Certain embodiments are therefore directed to a method for conditioning a medium. Other embodiments are directed to a medium that is conditioned. In one embodiment, the feeders are fibroblasts, and the medium is conditioned for approximately 24 hours. Other embodiments are directed to a method for transfecting a cell in vivo, wherein the transfection medium is conditioned. Other embodiments are directed to a method for reprogramming and/or gene-editing a cell in vivo, wherein the medium is conditioned. In one embodiment, the feeders are mitotically inactivated, for example, by exposure to a chemical such as mitomycin-C or by exposure to gamma radiation. In certain embodiments, it may be beneficial to use only autologous materials, in part to, for example and not wishing to be bound by theory, avoid the risk of disease transmission from the feeders to the cell or the patient. Certain embodiments are therefore directed to a method for transfecting a cell in vivo, wherein the transfection medium is conditioned, and wherein the feeders are derived from the same individual as the cell being transfected. Other embodiments are directed to a method for reprogramming and/or gene-editing a cell in vivo, wherein the medium is conditioned, and wherein the feeders are derived from the same individual as the cell being reprogrammed and/or gene-edited.
  • Several molecules can be added to media by conditioning. Certain embodiments are therefore directed to a medium that is supplemented with one or more molecules that are present in a conditioned medium. In one embodiment, the medium is supplemented with Wnt1, Wnt2, Wnt3, Wnt3a or a biologically active fragment, analogue, variant, agonist, or family-member thereof. In another embodiment, the medium is supplemented with TGF-β or a biologically active fragment, analogue, variant, agonist, or family-member thereof. In yet another embodiment, a cell in vivo is reprogrammed according to the method of the present invention, wherein the medium is not supplemented with TGF-β for between about 1 and about 5 days, and is then supplemented with TGF-β for at least about 2 days. In yet another embodiment, the medium is supplemented with IL-6, IL-6R or a biologically active fragment, analogue, variant, agonist, or family-member thereof. In yet another embodiment, the medium is supplemented with a sphingolipid or a fatty acid. In still another embodiment, the sphingolipid is lysophosphatidic acid, lysosphingomyelin, sphingosine-1-phosphate or a biologically active analogue, variant or derivative thereof.
  • In addition to mitotically inactivating cells, under certain conditions, irradiation can change the gene expression of cells, causing cells to produce less of certain proteins and more of certain other proteins that non-irradiated cells, for example, members of the Wnt family of proteins. In addition, certain members of the Wnt family of proteins can promote the growth and transformation of cells. It has now been discovered that, in certain situations, the efficiency of reprogramming can be greatly increased by contacting a cell in vivo with a medium that is conditioned using irradiated feeders instead of mitomycin-c-treated feeders. It has been further discovered that the increase in reprogramming efficiency observed when using irradiated feeders is caused in part by Wnt proteins that are secreted by the feeders. Certain embodiments are therefore directed to a method for reprogramming a cell in vivo, wherein the cell is contacted with Wnt1, Wnt2, Wnt3, Wnt3a or a biologically active fragment, analogue, variant, family-member or agonist thereof, including agonists of downstream targets of Wnt proteins, and/or agents that mimic one or more of the biological effects of Wnt proteins, for example, 2-amino-4-[3,4-(methylenedioxy)benzylamino]-6-(3-methoxyphenyl)pyrimidine.
  • Because of the low efficiency of many DNA-based reprogramming methods, these methods may be difficult or impossible to use with cells derived from patient samples, which may contain only a small number of cells. In contrast, the high efficiency of certain embodiments of the present invention can allow reliable reprogramming of a small number of cells, including single cells. Certain embodiments are directed to a method for reprogramming a small number of cells. Other embodiments are directed to a method for reprogramming a single cell. In one embodiment, the cell is contacted with one or more enzymes. In another embodiment, the enzyme is collagenase. In yet another embodiment, the collagenase is animal-component free. In one embodiment, the collagenase is present at a concentration of between about 0.1 mg/mL and about 10 mg/mL, or between about 0.5 mg/mL and about 5 mg/mL. In another embodiment, the cell is a blood cell. In yet another embodiment, the cell is contacted with a medium containing one or more proteins that is derived from the patient's blood. In still another embodiment, the cell is contacted with a medium comprising: DMEM/F12+2 mM L-alanyl-L-glutamine+between about 5% and about 25% patient-derived serum, or between about 10% and about 20% patient-derived serum, or about 20% patient-derived serum.
  • It has now been discovered that, in certain situations, transfecting cells in vivo with a mixture of RNA encoding Oct4, Sox2, Klf4, and c-Myc using the medium of the present invention can cause the rate of proliferation of the cells to increase. When the amount of RNA delivered to the cells is too low to ensure that all of the cells are transfected, only a fraction of the cells may show an increased proliferation rate. In certain situations, such as when generating a personalized therapeutic, increasing the proliferation rate of cells may be desirable, in part because doing so can reduce the time necessary to generate the therapeutic, and therefore can reduce the cost of the therapeutic. Certain embodiments are therefore directed to a method for transfecting a cell in vivo with a mixture of RNA encoding Oct4, Sox2, Klf4, and c-Myc. In one embodiment, the cell exhibits an increased proliferation rate. In another embodiment, the cell is reprogrammed.
  • Many diseases are associated with one or more mutations. Mutations can be corrected by contacting a cell with a nucleic acid that encodes a protein that, either alone or in combination with other molecules, corrects the mutation (an example of gene-editing). Examples of such proteins include: a nuclease, a transcription activator-like effector nuclease (TALEN), a zinc-finger nuclease, a meganuclease, a nickase, a clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof. Certain embodiments are therefore directed to a method for transfecting a cell in vivo with a nucleic acid, wherein the nucleic acid encodes a protein that, either alone or in combination with other molecules, creates a single-strand or double-strand break in a DNA molecule.
  • In a one embodiment, the protein is a zinc finger nuclease or a TALEN. In another embodiment, the nucleic acid is an RNA molecule. In yet another embodiment, the single-strand or double-strand break is within about 5,000,000 bases of the transcription start site of a gene selected from the group: CCR5, CXCR4, GAD1, GAD2, CFTR, HBA1, HBA2, HBB, HBD, FANCA, XPA, XPB, XPC, ERCC2, POLH, HTT, DMD, SOD1, APOE, PRNP, BRCA1, and BRCA2 or an analogue, variant or family-member thereof. In one embodiment, the present invention relates to gene-editing of the MYC protein (e.g. correcting one or more muations that may be linked to cancer), optionally with a TALEN. In yet another embodiment, the cell is transfected with a nucleic acid that acts as a repair template by either causing the insertion of a DNA sequence in the region of the single-strand or double-strand break or by causing the DNA sequence in the region of the single-strand or double-strand break to otherwise change. In yet another embodiment, the cell is reprogrammed, and subsequently, the cell is gene-edited. In yet another embodiment, the cell is gene-edited, and subsequently, the cell is reprogrammed. In yet another embodiment, the gene-editing and reprogramming are performed within about 7 days of each other. In yet another embodiment, the gene-editing and reprogramming occur simultaneously or on the same day. In yet another embodiment, the cell is a skin cell, the skin cell is gene-edited to disrupt the CCR5 gene, the skin cell is reprogrammed to a hematopoietic stem cell, thus producing a therapeutic for HIV/AIDS, and the therapeutic is used to treat a patient with HIV/AIDS. In yet another embodiment, the skin cell is derived from the same patient whom the therapeutic is used to treat.
  • Genes that can be edited according to the methods of the present invention to produce therapeutics of the present invention include genes that can be edited to restore normal function, as well as genes that can be edited to reduce or eliminate function. Such genes include, but are not limited to beta globin (HBB), mutations in which can cause sickle cell disease (SCD) and β-thalassemia, breast cancer 1, early onset (BRCA1) and breast cancer 2, early onset (BRCA2), mutations in which can increase susceptibility to breast cancer, C-C chemokine receptor type 5 (CCR5) and C-X-C chemokine receptor type 4 (CXCR4), mutations in which can confer resistance to HIV infection, cystic fibrosis transmembrane conductance regulator (CFTR), mutations in which can cause cystic fibrosis, dystrophin (DMD), mutations in which can cause muscular dystrophy, including Duchenne muscular dystrophy and Becker's muscular dystrophy, glutamate decarboxylase 1 and glutamate decarboxylase 2 (GAD1, GAD2), mutations in which can prevent autoimmune destruction of β-cells, hemoglobin alpha 1, hemoglobin alpha 2, and hemoglobin delta (HBA1, HBA2, and HBD), mutations in which can cause thalassemia, desmoplakin, keratin 5, keratin 14, plectin, integrin alpha-6, integrin beta-4, laminin subunit alpha-3, laminin subunit beta-3, laminin subunit gamma-2, collagen type VII alpha 1, collagen type XVII alpha 1, and matrix metalloproteinase-1 (DSP, KRT5, KRT14, PLEC1, ITGA6, ITGB4, LAMA3, LAMB3, LAMC2, COL7A1, COL17A1, and MMP1), mutations in which can cause epidermolysis bullosa, Huntington (HTT), mutations in which can cause Huntington's disease, superoxide dismutase 1 (SOD1), mutations in which can cause amyotrophic lateral sclerosis (ALS), XPA, XPB, XPC, XPD (ERCC6) and polymerase (DNA directed), eta (POLH), mutations in which can cause xeroderma pigmentosum, leucine-rich repeat kinase 2 (LRRK2), mutations in which can cause Parkinson's disease, and Fanconi anemia, complementation groups A, B, C, D1, D2, E, F, G, I, J, L, M, N, P (FANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ, FANCL, FANCM, FANCN, FANCP), and RAD51 homolog C (S. cerevisiae) (RAD51C), mutations in which can cause Fanconi anemia.
  • Certain embodiments are directed to a therapeutic comprising a nucleic acid. In one embodiment, the nucleic acid encodes one or more gene-editing proteins. Other embodiments are directed to a therapeutic comprising one or more cells that are transfected, reprogrammed, and/or gene-edited in vivo according to the methods of the present invention. In one embodiment, a cell is transfected, reprogrammed, and/or gene-edited, and the transfected, reprogrammed, and/or gene-edited cell is introduced into a patient. In another embodiment, the cell is harvested from the same patient into whom the transfected, reprogrammed and/or gene-edited cell is introduced. Examples of diseases that can be treated with therapeutics of the present invention include, but are not limited to Alzheimer's disease, spinal cord injury, amyotrophic lateral sclerosis, cystic fibrosis, heart disease, including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, Huntington's disease, diabetes, sickle-cell anemia, thalassemia, Fanconi anemia, xeroderma pigmentosum, muscular dystrophy, severe combined immunodeficiency, hereditary sensory neuropathy, cancer, and HIV/AIDS. In certain embodiments, the therapeutic comprises a cosmetic. In one embodiment, a cell is harvested from a patient, the cell is reprogrammed and expanded to a large number of adipose cells to produce a cosmetic, and the cosmetic is introduced into the patient. In still another embodiment, the cosmetic is used for tissue reconstruction.
  • While detailed examples are provided herein for the production of specific types of cells and for the production of therapeutics comprising specific types of cells, it is recognized that the methods of the present invention can be used to produce many other types of cells, and to produce therapeutics comprising one or more of many other types of cells, for example, by reprogramming a cell according to the methods of the present invention, and culturing the cell under conditions that mimic one or more aspects of development by providing conditions that resemble the conditions present in the cellular microenvironment during development.
  • Certain embodiments are directed to a library of cells with a variety of human leukocyte antigen (HLA) types (“HLA-matched libraries”). An HLA-matched library may be beneficial in part because it can provide for the rapid production and/or distribution of therapeutics without the patient having to wait for a therapeutic to be produced from the patient's cells. Such a library may be particularly beneficial for the production of cosmetics and for the treatment of heart disease and diseases of the blood and/or immune system for which patients may benefit from the immediate availability of a therapeutic or cosmetic.
  • The DNA sequence of a cell can be altered by contacting the cell with a gene-editing protein or by inducing the cell to express a gene-editing protein. However, previously disclosed gene-editing proteins suffer from low binding efficiency and excessive off-target activity, which can introduce undesired mutations in the DNA of the cell, severely limiting their use in vivo, for example in therapeutic and cosmetic applications, in which the introduction of undesired mutations in a patient's cells could lead to the development of cancer. It has now been discovered that gene-editing proteins that comprise the StsI endonuclease cleavage domain (SEQ ID NO: 1) can exhibit substantially lower off-target activity in vivo than previously disclosed gene-editing proteins, while maintaining a high level of on-target activity in vivo. Other novel engineered proteins have also been discovered that can exhibit high on-target activity in vivo, low off-target activity in vivo, small size, solubility, and other desirable characteristics when they are used as the nuclease domain of a gene-editing protein: StsI-HA (SEQ ID NO: 2), StsI-HA2 (SEQ ID NO: 3), StsI-UHA (SEQ ID NO: 4), StsI-UHA2 (SEQ ID NO: 5), StsI-HF (SEQ ID NO: 6), and StsI-UHF (SEQ ID NO: 7). StsI-HA, StsI-HA2 (high activity), StsI-UHA, and StsI-UHA2 (ultra-high activity) can exhibit higher on-target activity in vivo than both wild-type StsI and wild-type FokI, due in part to specific amino-acid substitutions within the N-terminal region at the 34 and 61 positions, while StsI-HF (high fidelity) and StsI-UHF (ultra-high fidelity) can exhibit lower off-target activity in vivo than both wild-type StsI and wild-type FokI, due in part to specific amino-acid substitutions within the C-terminal region at the 141 and 152 positions.
  • Certain embodiments are therefore directed to a protein. In some embodiments, the protein is present in vivo. In other embodiments, the protein comprises a nuclease domain. In one embodiment, the nuclease domain comprises one or more of: the cleavage domain of FokI endonuclease (SEQ ID NO: 53), the cleavage domain of StsI endonuclease (SEQ ID NO: 1), StsI-HA (SEQ ID NO: 2), StsI-HA2 (SEQ ID NO: 3), StsI-UHA (SEQ ID NO: 4), StsI-UHA2 (SEQ ID NO: 5), StsI-HF (SEQ ID NO: 6), and StsI-UHF (SEQ ID NO: 7) or a biologically active fragment or variant thereof.
  • It has also been discovered that engineered gene-editing proteins that comprise DNA-binding domains comprising certain novel repeat sequences can exhibit lower off-target activity in vivo than previously disclosed gene-editing proteins, while maintaining a high level of on-target activity in vivo. Certain of these engineered gene-editing proteins can provide several advantages over previously disclosed gene-editing proteins, including, for example, increased flexibility of the linker region connecting repeat sequences, which can result in increased binding efficiency. Certain embodiments are therefore directed to a protein comprising a plurality of repeat sequences. In one embodiment, at least one of the repeat sequences contains the amino-acid sequence: GabG, where “a” and “b” each represent any amino acid. In one embodiment, the protein is a gene-editing protein. In another embodiment, one or more of the repeat sequences are present in a DNA-binding domain. In a further embodiment, “a” and “b” are each independently selected from the group: H and G. In a still further embodiment, “a” and “b” are H and G, respectively. In one embodiment, the amino-acid sequence is present within about 5 amino acids of the C-terminus of the repeat sequence. In another embodiment, the amino-acid sequence is present at the C-terminus of the repeat sequence. In some embodiments, one or more G in the amino-acid sequence GabG is replaced with one or more amino acids other than G, for example A, H or GG. In one embodiment, the repeat sequence has a length of between about 32 and about 40 amino acids or between about 33 and about 39 amino acids or between about 34 and 38 amino acids or between about 35 and about 37 amino acids or about 36 amino acids or greater than about 32 amino acids or greater than about 33 amino acids or greater than about 34 amino acids or greater than about 35 amino acids. Other embodiments are directed to a protein comprising one or more transcription activator-like effector domains. In one embodiment, at least one of the transcription activator-like effector domains comprises a repeat sequence. Other embodiments are directed to a protein comprising a plurality of repeat sequences generated by inserting one or more amino acids between at least two of the repeat sequences of a transcription activator-like effector domain. In one embodiment, one or more amino acids is inserted about 1 or about 2 or about 3 or about 4 or about 5 amino acids from the C-terminus of at least one repeat sequence. Still other embodiments are directed to a protein comprising a plurality of repeat sequences, wherein about every other repeat sequence has a different length than the repeat sequence immediately preceding or following the repeat sequence. In one embodiment, every other repeat sequence is about 36 amino acids long. In another embodiment, every other repeat sequence is 36 amino acids long. Still other embodiments are directed to a protein comprising a plurality of repeat sequences, wherein the plurality of repeat sequences comprises at least two repeat sequences that are each at least 36 amino acids long, and wherein at least two of the repeat sequences that are at least 36 amino acids long are separated by at least one repeat sequence that is less than 36 amino acids long. Some embodiments are directed to a protein that comprises one or more sequences selected from, for example, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, and SEQ ID NO: 60.
  • Other embodiments are directed to a protein that comprises a DNA-binding domain. In some embodiments, the DNA-binding domain comprises a plurality of repeat sequences. In one embodiment, the plurality of repeat sequences enables high-specificity recognition of a binding site in a target DNA molecule. In another embodiment, at least two of the repeat sequences have at least about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 95%, or about 98%, or about 99% homology to each other. In a further embodiment, at least one of the repeat sequences comprises one or more regions capable of binding to a binding site in a target DNA molecule. In a still further embodiment, the binding site comprises a defined sequence of between about 1 to about 5 bases in length. In one embodiment, the DNA-binding domain comprises a zinc finger. In another embodiment, the DNA-binding domain comprises a transcription activator-like effector (TALE). In a further embodiment, the plurality of repeat sequences includes at least one repeat sequence having at least about 50% or about 60% or about 70% or about 80% or about 90% or about 95% or about 98%, or about 99% homology to a TALE. In a still further embodiment, the gene-editing protein comprises a clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein. In one embodiment, the gene-editing protein comprises a nuclear-localization sequence. In another embodiment, the nuclear-localization sequence comprises the amino-acid sequence: PKKKRKV (SEQ ID NO:471). In one embodiment, the gene-editing protein comprises a mitochondrial-localization sequence. In another embodiment, the mitochondrial-localization sequence comprises the amino-acid sequence: LGRVIPRKIASRASLM (SEQ ID NO:472). In one embodiment, the gene-editing protein comprises a linker. In another embodiment, the linker connects a DNA-binding domain to a nuclease domain. In a further embodiment, the linker is between about 1 and about 10 amino acids long. In some embodiments, the linker is about 1, about 2, or about 3, or about 4, or about 5, or about 6, or about 7, or about 8, or about 9, or about 10 amino acids long. In one embodiment, the gene-editing protein is capable of generating a nick or a double-strand break in a target DNA molecule.
  • Certain embodiments are directed to a method for modifying the genome of a cell in vivo, the method comprising introducing into a cell in vivo a nucleic acid molecule encoding a non-naturally occurring fusion protein comprising an artificial transcription activator-like (TAL) effector repeat domain comprising one or more repeat units 36 amino acids in length and an endonuclease domain, wherein the repeat domain is engineered for recognition of a predetermined nucleotide sequence, and wherein the fusion protein recognizes the predetermined nucleotide sequence. In one embodiment, the cell is a eukaryotic cell. In another embodiment, the cell is an animal cell. In a further embodiment, the cell is a mammalian cell. In a still further embodiment, the cell is a human cell. In one embodiment, the cell is a plant cell. In another embodiment, the cell is a prokaryotic cell. In some embodiments, the fusion protein introduces an endonucleolytic cleavage in a nucleic acid of the cell, whereby the genome of the cell is modified.
  • Certain embodiments are directed to a composition for altering the DNA sequence of a cell in vivo comprising a nucleic acid, wherein the nucleic acid encodes a gene-editing protein. Other embodiments are directed to a composition for altering the DNA sequence of a cell in vivo comprising a nucleic-acid mixture, wherein the nucleic-acid mixture comprises: a first nucleic acid that encodes a first gene-editing protein, and a second nucleic acid that encodes a second gene-editing protein. In one embodiment, the binding site of the first gene-editing protein and the binding site of the second gene-editing protein are present in the same target DNA molecule. In another embodiment, the binding site of the first gene-editing protein and the binding site of the second gene-editing protein are separated by less than about 50 bases, or less than about 40 bases, or less than about 30 bases or less than about 20 bases, or less than about 10 bases, or between about 10 bases and about 25 bases or about 15 bases. In one embodiment, the nuclease domain of the first gene-editing protein and the nuclease domain of the second gene-editing protein are capable of forming a dimer. In another embodiment, the dimer is capable of generating a nick or double-strand break in a target DNA molecule.
  • Certain embodiments are directed to a therapeutic composition. Other embodiments are directed to a cosmetic composition. In some embodiments, the composition comprises a repair template. In a further embodiment, the repair template is a single-stranded DNA molecule or a double-stranded DNA molecule.
  • Other embodiments are directed to an article of manufacture for synthesizing a protein or a nucleic acid encoding a protein. In one embodiment, the article is a nucleic acid. In another embodiment, the protein comprises a DNA-binding domain. In a further embodiment, the nucleic acid comprises a nucleotide sequence encoding a DNA-binding domain. In one embodiment, the protein comprises a nuclease domain. In another embodiment, the nucleic acid comprises a nucleotide sequence encoding a nuclease domain. In one embodiment, the protein comprises a plurality of repeat sequences. In another embodiment, the nucleic acid encodes a plurality of repeat sequences. In a further embodiment, the nuclease domain is selected from: FokI, StsI, StsI-HA, StsI-HA2, StsI-UHA, StsI-UHA2, StsI-HF, and StsI-UHF or a natural or engineered variant or biologically active fragment thereof. In one embodiment, the nucleic acid comprises an RNA-polymerase promoter. In another embodiment, the RNA-polymerase promoter is a T7 promoter or a SP6 promoter. In a further embodiment, the nucleic acid comprises a viral promoter. In one embodiment, the nucleic acid comprises an untranslated region. In another embodiment, the nucleic acid is an in vitro-transcription template.
  • Certain embodiments are directed to a method for inducing a cell to express a protein in vivo. Other embodiments are directed to a method for altering the DNA sequence of a cell in vivo comprising transfecting the cell in vivo with a gene-editing protein or inducing the cell to express a gene-editing protein in vivo. Still other embodiments are directed to a method for reducing the expression of a protein of interest in a cell in vivo. In one embodiment, the cell is induced to express a gene-editing protein, wherein the gene-editing protein is capable of creating a nick or a double-strand break in a target DNA molecule. In another embodiment, the nick or double-strand break results in inactivation of a gene. Still other embodiments are directed to a method for generating an inactive, reduced-activity or dominant-negative form of a protein in vivo. In one embodiment, the protein is survivin. Still other embodiments are directed to a method for repairing one or more mutations in a cell in vivo. In one embodiment, the cell is contacted with a repair template. In another embodiment, the repair template is a DNA molecule. In a further embodiment, the repair template does not contain a binding site of the gene-editing protein. In a still further embodiment, the repair template encodes an amino-acid sequence that is encoded by a DNA sequence that comprises a binding site of the gene-editing protein.
  • In various embodiments, the repair template is about 20 nucleotides, or about 30 nucleotides, or about 40 nucleotides, or about 50 nucleotides, or about 60 nucleotides, or about 70 nucleotides, or about 80 nucleotides, or about 90 nucleotides, or about 100 nucleotides, or about 150 nucleotides, or about 200 nucleotides, or about 300 nucleotides, or about 400 nucleotides, or about 500 nucleotides, or about 750 nucleotides, or about 1000 nucleotides. In various embodiments, the repair template is about 20-1000 nucleotides, or about 20-500 nucleotides, or about 20-400 nucleotides, or about 20-200 nucleotides, or about 20-100 nucleotides, or about 80-100 nucleotides, or about 50-100 nucleotides.
  • In various embodiments, the mass ratio of RNA (e.g. synthetic RNA encoding gene-editing protein) to repair template is about 1:10, or about 1:9, or about 1:8, or about 1:7, or about 1:6, or about 1:5, or about 1:4, or about 1:3, or about 1:2, or about 1:1, or about 2:1, or about 3:1, or about 4:1, or about 5:1, or about 6:1, or about 7:1, or about 8:1, or about 9:1, or about 10:1.
  • In various embodiments, the molar ratio of RNA (e.g. synthetic RNA encoding gene-editing protein) to repair template is about 1:10, or about 1:9, or about 1:8, or about 1:7, or about 1:6, or about 1:5, or about 1:4, or about 1:3, or about 1:2, or about 1:1, or about 2:1, or about 3:1, or about 4:1, or about 5:1, or about 6:1, or about 7:1, or about 8:1, or about 9:1, or about 10:1.
  • In various embodiments, the repair template has a dual function, causing a repair to a gene-edited target sequence and preventing further binding of a gene-editing protein, thereby reducing or eliminating further gene-editing (e.g. via the repair template causing a repair that renders what was the gene-editing protein binding site no longer suitable for gene-editing protein binding). Accordingly, in some embodiments, the present gene-editing methods are tunable to ensure a single gene-edit per target site.
  • Other embodiments are directed to a method for treating a patient comprising administering to the patient a therapeutically or cosmetically effective amount of a protein or a nucleic acid encoding a protein. In one embodiment, the treatment results in one or more of the patient's symptoms being ameliorated. Certain embodiments are directed to a method for treating a patient comprising: a. inducing a cell to express a protein of interest by transfecting the cell in vivo with a nucleic acid encoding the protein of interest and/or b. reprogramming the cell in vivo. In one embodiment, the cell is reprogrammed to a less differentiated state. In another embodiment, the cell is reprogrammed by transfecting the cell with one or more synthetic RNA molecules encoding one or more reprogramming proteins. In a further embodiment, the cell is differentiated. In a still further embodiment, the cell is differentiated into one of: a skin cell, a glucose-responsive insulin-producing cell, a hematopoietic cell, a cardiac cell, a retinal cell, a renal cell, a neural cell, a stromal cell, a fat cell, a bone cell, a muscle cell, an oocyte, and a sperm cell. Other embodiments are directed to a method for treating a patient comprising: a. inducing a cell to express a gene-editing protein by transfecting the cell in vivo with a nucleic acid encoding a gene-editing protein and/or b. reprogramming the cell in vivo.
  • Other embodiments are directed to a complexation medium. In one embodiment, the complexation medium has a pH greater than about 7, or greater than about 7.2, or greater than about 7.4, or greater than about 7.6, or greater than about 7.8, or greater than about 8.0, or greater than about 8.2, or greater than about 8.4, or greater than about 8.6, or greater than about 8.8, or greater than about 9.0. In another embodiment, the complexation medium comprises transferrin. In a further embodiment, the complexation medium comprises DMEM. In a still further embodiment, the complexation medium comprises DMEM/F12. Still other embodiments are directed to a method for forming nucleic-acid-transfection-reagent complexes. In one embodiment, the transfection reagent is incubated with a complexation medium. In another embodiment, the incubation occurs before a mixing step. In a further embodiment, the incubation step is between about 5 seconds and about 5 minutes or between about 10 seconds and about 2 minutes or between about 15 seconds and about 1 minute or between about 30 seconds and about 45 seconds. In one embodiment, the transfection reagent is selected from Table 1. In another embodiment, the transfection reagent is a lipid or lipidoid. In a further embodiment, the transfection reagent comprises a cation. In a still further embodiment, the cation is a multivalent cation. In a still further embodiment, the transfection reagent is N1-[2-((1S)-1-[(3-aminopropyl)amino]-4-[di(3-amino-propyl)amino]butylcarboxamido)ethyl]-3,4-di[oleyloxy]-benzamide (a.k.a. MVL5) or a derivative thereof.
  • Certain embodiments are directed to a method for inducing a cell to express a protein by contacting the cell with a nucleic acid in vivo. In one embodiment, the cell is a mammalian cell. In another embodiment, the cell is a human cell or a rodent cell. Other embodiments are directed to a cell produced using one or more of the methods of the present invention. In one embodiment, the cell is present in a patient. In another embodiment, the cell is isolated from a patient. Other embodiments are directed to a screening library comprising a cell produced using one or more of the methods of the present invention. In one embodiment, the screening library is used for at least one of: toxicity screening, including: cardiotoxicity screening, neurotoxicity screening, and hepatotoxicity screening, efficacy screening, high-throughput screening, high-content screening, and other screening.
  • Other embodiments are directed to a kit containing a nucleic acid. In one embodiment, the kit contains a delivery reagent (a.k.a. “transfection reagent”). In another embodiment, the kit is a reprogramming kit. In a further embodiment, the kit is a gene-editing kit. Other embodiments are directed to a kit for producing nucleic acids. In one embodiment, the kit contains at least two of: pseudouridine-triphosphate, 5-methyluridine triphosphate, 5-methylcytidine triphosphate, 5-hydroxymethylcytidine triphosphate, N4-methylcytidine triphosphate, N4-acetylcytidine triphosphate, and 7-deazaguanosine triphosphate or one or more derivatives thereof. Other embodiments are directed to a therapeutic or cosmetic comprising a nucleic acid. In one embodiment, the therapeutic or cosmetic is a pharmaceutical composition. In another embodiment, the pharmaceutical composition is formulated. In a further embodiment, the formulation comprises an aqueous suspension of liposomes. Example liposome components are set forth in Table 1, and are given by way of example, and not by way of limitation. In one embodiment, the liposomes include one or more polyethylene glycol (PEG) chains. In another embodiment, the PEG is PEG2000. In a further embodiment, the liposomes include 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) or a derivative thereof. In one embodiment, the therapeutic comprises one or more ligands. In another embodiment, the therapeutic comprises at least one of: androgen, CD30 (TNFRSF8), a cell-penetrating peptide, CXCR, estrogen, epidermal growth factor, EGFR, HER2, folate, insulin, insulin-like growth factor-I, interleukin-13, integrin, progesterone, stromal-derived-factor-1, thrombin, vitamin D, and transferrin or a biologically active fragment or variant thereof. Still other embodiments are directed to a therapeutic or cosmetic comprising a cell generated using one or more of the methods of the present invention.
  • In one embodiment, the therapeutic is administered to a patient for the treatment of any of the diseases or disorders described herein, including by way of non-limitation, type 1 diabetes, heart disease, including ischemic and dilated cardiomyopathy, macular degeneration, Parkinson's disease, cystic fibrosis, sickle-cell anemia, thalassemia, Fanconi anemia, severe combined immunodeficiency, hereditary sensory neuropathy, xeroderma pigmentosum, Huntington's disease, muscular dystrophy, amyotrophic lateral sclerosis, Alzheimer's disease, cancer, and infectious diseases including: hepatitis and HIV/AIDS.
  • Other embodiments are directed to a method for reprogramming a cell in vivo. In one embodiment, the cell is reprogrammed by contacting the cell with one or more nucleic acids. In one embodiment, the cell is contacted with a plurality of nucleic acids encoding at least one of: Oct4 protein, Sox2 protein, Klf4 protein, c-Myc protein, Lin28 protein or a biologically active fragment, variant or derivative thereof. In another embodiment, the cell is contacted with a plurality of nucleic acids encoding a plurality of proteins including: Oct4 protein, Sox2 protein, Klf4 protein, and c-Myc protein or one or more biologically active fragments, variants or derivatives thereof. Still other embodiments are directed to a method for gene editing a cell in vivo. In one embodiment, the cell is gene-edited by contacting the cell with one or more nucleic acids.
  • Certain embodiments are directed to a method for inducing a cell in vivo to express a protein of interest comprising contacting a cell in vivo with a solution comprising albumin that is treated with an ion-exchange resin or charcoal and one or more nucleic acid molecules, wherein at least one of the one or more nucleic acid molecules encodes a protein of interest. In one embodiment, the method results in the cell expressing the protein of interest. In another embodiment, the one or more nucleic acid molecules comprise a synthetic RNA molecule. In one embodiment, the cell is a skin cell. In another embodiment, the cell is a muscle cell. In yet another embodiment, the cell is a dermal fibroblast. In yet another embodiment, the cell is a myoblast. In one embodiment, the protein of interest is an extracellular matrix protein. In another embodiment, the protein of interest is selected from: elastin, collagen, laminin, fibronectin, vitronectin, lysyl oxidase, elastin binding protein, a growth factor, fibroblast growth factor, transforming growth factor beta, granulocyte colony-stimulating factor, a matrix metalloproteinase, an actin, fibrillin, microfibril-associated glycoprotein, a lysyl-oxidase-like protein, and platelet-derived growth factor. In one embodiment, the solution is delivered to the dermis. In another embodiment, the delivering is by injection. In yet another embodiment, the delivering is by injection using a microneedle array. In one embodiment, the solution further comprises a growth factor. In another embodiment, the growth factor is selected from: fibroblast growth factor and transforming growth factor beta. In yet another embodiment, the solution further comprises cholesterol. Other embodiments are directed a method for inducing a cell in vivo to express a protein of interest comprising contacting a cell in vivo with a solution comprising cholesterol and one or more nucleic acid molecules, wherein at least one of the one or more nucleic acid molecules encodes a protein of interest. In one embodiment, the method results in the cell expressing the protein of interest. Still other embodiments are directed to a method for transfecting a cell in vivo with a nucleic acid molecule comprising contacting a cell in vivo with a solution comprising albumin that is treated with an ion-exchange resin or charcoal and a nucleic acid molecule. In one embodiment, the method results in the cell being transfected with the nucleic acid molecule. In another embodiment, the nucleic acid molecule is one of: a dsDNA molecule, a ssDNA molecule, a dsRNA molecule, a ssRNA molecule, a plasmid, an oligonucleotide, a synthetic RNA molecule, a miRNA molecule, an mRNA molecule, an siRNA molecule. Still other embodiments are directed to a method for treating a patient comprising delivering to a patient a composition comprising albumin that is treated with an ion-exchange resin or charcoal and one or more nucleic acid molecules, wherein at least one of the one or more nucleic acid molecules encodes a protein of interest. In one embodiment, the method results in the expression of the protein of interest in the patient. In another embodiment, the method results in the expression of the protein of interest in the dermis of the patient.
  • Certain embodiments are directed to a cosmetic composition comprising albumin that is treated with an ion-exchange resin or charcoal and a nucleic acid molecule. Other embodiments are directed to a cosmetic treatment article. In one embodiment, the cosmetic treatment article comprises a device configured to deliver a composition to a patient. In another embodiment, the nucleic acid molecule encodes elastin protein or collagen protein. Still other embodiments are directed to a solution for transfecting a cell in vivo comprising cholesterol or a cholesterol analog and one or more nucleic acid molecules. In one embodiment, the cholesterol or cholesterol analog is covalently bound to at least one of the one or more nucleic acid molecules. In another embodiment, the cholesterol analog is an oxysterol. In yet another embodiment, the cholesterol analog includes one or more of: an A-ring substitution, a B-ring substitution, a D-ring substitution, a side-chain substitution, a cholestanoic acid, a cholestenoic acid, a polyunsaturated moiety, a deuterated moiety, a fluorinated moiety, a sulfonated moiety, a phosphorylated moiety, and a fluorescent moiety. In yet another embodiment, the method comprises treating the patient with one or more of: a dermal filler, a neurotoxin (by way of illustration sodium channel inhibitors (e.g., tetrodotoxin), potassium channel inhibitors (e.g., tetraethylammonium), chloride channel inhibitors (e.g., chlorotoxin and curare), calcium channel inhibitors (e.g., conotoxin), synaptic vesicle release inhibitors (e.g., botulinum toxin and tetanus toxin) and blood brain barrier inhibitor (e.g., aluminum and mercury)) and a repair-inducing treatment.
  • Despite the tendency of transfection reagent nucleic acid complexes to precipitate, form clumps or otherwise degrade when stored for more than a few minutes, the present inventors have surprisingly discovered that transfection reagent nucleic acid complexes produced according to some embodiments of the present invention can be frozen and/or can be stored at various temperatures, including room temperature, about 4° C., about −20° C., about −80° C., and about −196° C. for an extended period of time, for example, for several hours, about 1 day, about 1 week, about 1 month, about 1 year, and longer than about 1 year. Some embodiments are therefore directed to a pharmaceutical formulation comprising synthetic RNA and a transfection reagent, wherein the pharmaceutical formulation is provided in solid form. Other embodiments are directed to a pharmaceutical formulation comprising synthetic RNA transfection reagent complexes, wherein the synthetic RNA transfection reagent complexes are provided in solid form. In various embodiments, the synthetic RNA transfection reagent complexes are provided in frozen form. Various embodiments are directed to a method for stabilizing nucleic acid transfection reagent complexes comprising forming nucleic acid transfection reagent complexes and contacting the nucleic acid transfection reagent complexes or vessel in which such are contained with a cryogenic liquid to produce stabilized nucleic acid transfection reagent complexes. In one embodiment, the nucleic acid transfection reagent complexes are stabilized for shipment or storage.
  • Illustrative subjects or patients refers to any vertebrate including, without limitation, humans and other primates (e.g., chimpanzees and other apes and monkey species), farm animals (e.g., cattle, sheep, pigs, goats, and horses), domestic mammals (e.g., dogs and cats), laboratory animals (e.g., rodents such as mice, rats, and guinea pigs), and birds (e.g., domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like). In some embodiments, the subject is a mammal. In some embodiments, the subject is a human.
  • Definitions
  • By “molecule” is meant a molecular entity (molecule, ion, complex, etc.).
  • By “RNA molecule” is meant a molecule that comprises RNA.
  • By “synthetic RNA molecule” is meant an RNA molecule that is produced outside of a cell or that is produced inside of a cell using bioengineering, by way of non-limiting example, an RNA molecule that is produced in an in vitro-transcription reaction, an RNA molecule that is produced by direct chemical synthesis or an RNA molecule that is produced in a genetically-engineered E. coli cell.
  • By “transfection” is meant contacting a cell with a molecule, wherein the molecule is internalized by the cell.
  • By “upon transfection” is meant during or after transfection.
  • By “transfection reagent” is meant a substance or mixture of substances that associates with a molecule and facilitates the delivery of the molecule to and/or internalization of the molecule by a cell, by way of non-limiting example, a cationic lipid, a charged polymer or a cell-penetrating peptide.
  • By “reagent-based transfection” is meant transfection using a transfection reagent.
  • By “medium” is meant a solvent or a solution comprising a solvent and a solute, by way of non-limiting example, Dulbecco's Modified Eagle's Medium (DMEM), DMEM+10% fetal bovine serum (FBS), saline or water.
  • By “complexation medium” is meant a medium to which a transfection reagent and a molecule to be transfected are added and in which the transfection reagent associates with the molecule to be transfected.
  • By “transfection medium” is meant a medium that can be used for transfection, by way of non-limiting example, Dulbecco's Modified Eagle's Medium (DMEM), DMEM/F12, saline or water.
  • By “recombinant protein” is meant a protein or peptide that is not produced in animals or humans. Non-limiting examples include human transferrin that is produced in bacteria, human fibronectin that is produced in an in vitro culture of mouse cells, and human serum albumin that is produced in a rice plant.
  • By “Oct4 protein” is meant a protein that is encoded by the POU5F1 gene, or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof, by way of non-limiting example, human Oct4 protein (SEQ ID NO: 8), mouse Oct4 protein, Oct1 protein, a protein encoded by POU5F1 pseudogene 2, a DNA-binding domain of Oct4 protein or an Oct4-GFP fusion protein. In some embodiments the Oct4 protein comprises an amino acid sequence that has at least 70% identity with SEQ ID NO: 8, or in other embodiments, at least 75%, 80%, 85%, 90%, or 95% identity with SEQ ID NO: 8. In some embodiments, the Oct4 protein comprises an amino acid sequence having from 1 to 20 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 8. Or in other embodiments, the Oct4 protein comprises an amino acid sequence having from 1 to 15 or from 1 to 10 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 8.
  • By “Sox2 protein” is meant a protein that is encoded by the SOX2 gene, or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof, by way of non-limiting example, human Sox2 protein (SEQ ID NO: 9), mouse Sox2 protein, a DNA-binding domain of Sox2 protein or a Sox2-GFP fusion protein. In some embodiments the Sox2 protein comprises an amino acid sequence that has at least 70% identity with SEQ ID NO: 9, or in other embodiments, at least 75%, 80%, 85%, 90%, or 95% identity with SEQ ID NO: 9. In some embodiments, the Sox2 protein comprises an amino acid sequence having from 1 to 20 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 9. Or in other embodiments, the Sox2 protein comprises an amino acid sequence having from 1 to 15 or from 1 to 10 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 9.
  • By “Klf4 protein” is meant a protein that is encoded by the KLF4 gene, or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof, by way of non-limiting example, human Klf4 protein (SEQ ID NO: 10), mouse Klf4 protein, a DNA-binding domain of Klf4 protein or a Klf4-GFP fusion protein. In some embodiments the Klf4 protein comprises an amino acid sequence that has at least 70% identity with SEQ ID NO: 10, or in other embodiments, at least 75%, 80%, 85%, 90%, or 95% identity with SEQ ID NO: 10. In some embodiments, the Klf4 protein comprises an amino acid sequence having from 1 to 20 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 10. Or in other embodiments, the Klf4 protein comprises an amino acid sequence having from 1 to 15 or from 1 to 10 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 10.
  • By “c-Myc protein” is meant a protein that is encoded by the MYC gene, or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof, by way of non-limiting example, human c-Myc protein (SEQ ID NO: 11), mouse c-Myc protein, I-Myc protein, c-Myc (T58A) protein, a DNA-binding domain of c-Myc protein or a c-Myc-GFP fusion protein. In some embodiments the c-Myc protein comprises an amino acid sequence that has at least 70% identity with SEQ ID NO: 11, or in other embodiments, at least 75%, 80%, 85%, 90%, or 95% identity with SEQ ID NO: 11. In some embodiments, the c-Myc protein comprises an amino acid having from 1 to 20 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 11. Or in other embodiments, the c-Myc protein comprises an amino acid sequence having from 1 to 15 or from 1 to 10 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 11.
  • By “erythropoietin” or “erythropoietin protein” is meant a protein that is encoded by the EPO gene, or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof, by way of non-limiting example, human erythropoietin (SEQ ID NO: 164), mouse erythropoietin, darbepoetin, darbepoetin alfa, NOVEPOETIN, a binding domain of erythropoietin or an erythropoietin-GFP fusion protein. In some embodiments the erythropoietin comprises an amino acid sequence that has at least 70% identity with SEQ ID NO: 164, or in other embodiments, at least 75%, 80%, 85%, 90%, or 95% identity with SEQ ID NO: 164. In some embodiments, the erythropoietin comprises an amino acid sequence having from 1 to 20 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 164. Or in other embodiments, the erythropoietin comprises an amino acid sequence having from 1 to 15 or from 1 to 10 amino acid insertions, deletions, or substitutions (collectively) with respect to SEQ ID NO: 164.
  • By “reprogramming” is meant causing a change in the phenotype of a cell, by way of non-limiting example, causing a β-cell progenitor to differentiate into a mature β-cell, causing a fibroblast to dedifferentiate into a pluripotent stem cell, causing a keratinocyte to transdifferentiate into a cardiac stem cell, causing the telomeres of a cell to lengthen or causing the axon of a neuron to grow.
  • By “reprogramming factor” is meant a molecule that, when a cell is contacted with the molecule and/or the cell expresses the molecule, can, either alone or in combination with other molecules, cause reprogramming, by way of non-limiting example, Oct4 protein, Tert protein or erythropoietin.
  • By “germ cell” is meant a sperm cell or an egg cell.
  • By “pluripotent stem cell” is meant a cell that can differentiate into cells of all three germ layers (endoderm, mesoderm, and ectoderm) in vivo.
  • By “somatic cell” is meant a cell that is not a pluripotent stem cell or a germ cell, by way of non-limiting example, a skin cell.
  • By “hematopoietic cell” is meant a blood cell or a cell that can differentiate into a blood cell, by way of non-limiting example, a hematopoietic stem cell or a white blood cell.
  • By “cardiac cell” is meant a heart cell or a cell that can differentiate into a heart cell, by way of non-limiting example, a cardiac stem cell or a cardiomyocyte.
  • By “retinal cell” is meant a cell of the retina or a cell that can differentiate into a cell of the retina, by way of non-limiting example, a retinal pigmented epithelial cell.
  • By “skin cell” is meant a cell that is normally found in the skin, by way of non-limiting example, a fibroblast, a keratinocyte, a melanocyte, an adipocyte, a mesenchymal stem cell, an adipose stem cell or a blood cell.
  • By “immunosuppressant” is meant a substance that can suppress one or more aspects of an immune system, and that is not normally present in a mammal, by way of non-limiting example, B18R or dexamethasone.
  • By “single-strand break” is meant a region of single-stranded or double-stranded DNA in which one or more of the covalent bonds linking the nucleotides has been broken in one of the one or two strands.
  • By “double-strand break” is meant a region of double-stranded DNA in which one or more of the covalent bonds linking the nucleotides has been broken in each of the two strands.
  • By “nucleotide” is meant a nucleotide or a fragment or derivative thereof, by way of non-limiting example, a nucleobase, a nucleoside, a nucleotide-triphosphate, etc.
  • By “nucleoside” is meant a nucleotide or a fragment or derivative thereof, by way of non-limiting example, a nucleobase, a nucleoside, a nucleotide-triphosphate, etc.
  • By “gene editing” is meant altering the DNA sequence of a cell, by way of non-limiting example, by transfecting the cell with a protein that causes a mutation in the DNA of the cell.
  • By “gene-editing protein” is meant a protein that can, either alone or in combination with one or more other molecules, alter the DNA sequence of a cell, by way of non-limiting example, a nuclease, a transcription activator-like effector nuclease (TALEN), a zinc-finger nuclease, a meganuclease, a nickase, a clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein or a natural or engineered variant, family-member, orthologue, fragment or fusion construct thereof.
  • By “repair template” is meant a nucleic acid containing a region of at least about 70% homology with a sequence that is within 10 kb of a target site of a gene-editing protein.
  • By “repeat sequence” is meant an amino-acid sequence that is present in more than one copy in a protein, to within at least about 10% homology, by way of non-limiting example, a monomer repeat of a transcription activator-like effector.
  • By “DNA-binding domain” is meant a region of a molecule that is capable of binding to a DNA molecule, by way of non-limiting example, a protein domain comprising one or more zinc fingers, a protein domain comprising one or more transcription activator-like (TAL) effector repeat sequences or a binding pocket of a small molecule that is capable of binding to a DNA molecule.
  • By “binding site” is meant a nucleic-acid sequence that is capable of being recognized by a gene-editing protein, DNA-binding protein, DNA-binding domain or a biologically active fragment or variant thereof or a nucleic-acid sequence for which a gene-editing protein, DNA-binding protein, DNA-binding domain or a biologically active fragment or variant thereof has high affinity, by way of non-limiting example, an about 20-base-pair sequence of DNA in exon 1 of the human BIRC5 gene.
  • By “target” is meant a nucleic acid that contains a binding site.
  • Other definitions are set forth in U.S. application Ser. No. 13/465,490, U.S. Provisional Application No. 61/664,494, U.S. Provisional Application No. 61/721,302, International Application No. PCT/US12/67966, U.S. Provisional Application No. 61/785,404, U.S. Provisional Application No. 61/842,874, International Application No. PCT/US13/68118, U.S. Provisional Application No. 61/934,397, U.S. application Ser. No. 14/296,220, U.S. Provisional Application No. 62/038,608, U.S. Provisional Application No. 62/069,667, and International Application No. PCT/US2015/013949, the contents of which are hereby incorporated by reference in their entireties.
  • Selected Sequences
  • SEQ ID NO Description
    1 Stsl
    2 Stsl-HA
    3 Stsl-HA2
    4 Stsl-UHA
    5 Stsl-UHA2
    6 Stsl-HF
    7 Stsl-UHF
    8 Oct4
    9 Sox2
    10 Klf4
    11 c-Myc
    12 BIRC5_exon1
    13 BIRC5_exon2
    14 BIRC5_exon3
    15 BIRC5_exon4
    16 BIRC5-1.1-L
    17 BIRC5-1.1-R
    18 BIRC5-1.2-L
    19 BIRC5-1.2-R
    20 BIRC5-1.3-L
    21 BIRC5-1.3-R
    22 BIRC5-2.1-L
    23 BIRC5-2.1-R
    24 BIRC5-2.2-L
    25 BIRC5-2.2-R
    26 BIRC5-3.1-L
    27 BIRC5-3.1-R
    28 CDK1
    29 CDK2
    30 CDK3
    31 CDK4
    32 CDK5
    33 CDK6
    34 BIRC5
    35 HIF1A
    36 RRM2
    37 KRAS
    38 EGFR
    39 MYC
    40 PKN3
    41 KIF11
    42 APC
    43 BRCA1
    44 BRCA2
    45 TP53
    46 APP
    47 HTT
    48 IAPP
    49 MAPT
    50 PRNP
    51 SNCA
    52 SOD1
    53 Fokl
    54 Repeat1
    55 Repeat2
    56 Repeat3
    57 EO-GHGG-Fokl (“GHGG” disclosed as SEQ ID NO: 547)
    58 GHGG-Fokl (“GHGG” disclosed as SEQ ID NO: 547)
    59 EO-GHGG-Stsl (“GHGG” disclosed as SEQ ID NO: 547)
    60 GHGG-Stsl (“GHGG” disclosed as SEQ ID NO: 547)
    61 collagen alpha-1(I) chain preproprotein
    62 collagen alpha-2(I) chain precursor
    63 collagen alpha-1(II) chain isoform 1 precursor
    64 collagen alpha-1(II) chain isoform 2 precursor
    65 collagen alpha-1(III) chain preproprotein
    66 collagen alpha-1(IV) chain preproprotein
    67 collagen alpha-2(IV) chain preproprotein
    68 collagen alpha-3(IV) chain precursor
    69 collagen alpha-4(IV) chain precursor
    70 collagen alpha-5(IV) chain isoform 1 precursor
    71 collagen alpha-6(IV) chain isoform A precursor
    72 collagen alpha-1(V) chain isoform 1 preproprotein
    73 collagen alpha-2(V) chain preproprotein
    74 collagen alpha-3(V) chain preproprotein
    75 collagen alpha-1(VI) chain precursor
    76 collagen alpha-2(VI) chain isoform 2C2 precursor
    77 collagen alpha-3(VI) chain isoform 1 precursor
    78 collagen alpha-1(VII) chain precursor
    79 elastin isoform a precursor
    80 elastin isoform b precursor
    81 elastin isoform c precursor
    82 elastin isoform d precursor
    83 elastin isoform e precursor
    84 elastin isoform f precursor
    85 elastin isoform g precursor
    86 elastin isoform h precursor
    87 elastin isoform i precursor
    88 elastin isoform j precursor
    89 elastin isoform k precursor
    90 elastin isoform l precursor
    91 elastin isoform m precursor
    92 protein-lysine 6-oxidase isoform 1 preproprotein
    93 protein-lysine 6-oxidase isoform 2
    94 telomerase reverse transcriptase isoform 1
    95 telomerase reverse transcriptase isoform 2
    96 fibronectin isoform 1 preproprotein
    97 fibronectin isoform 3 preproprotein
    98 fibronectin isoform 4 preproprotein
    99 fibronectin isoform 5 preproprotein
    100 fibronectin isoform 6 preproprotein
    101 fibronectin isoform 7 preproprotein
    102 vitronectin precursor
    103 nidogen-1 precursor
    104 laminin subunit alpha-1 precursor
    105 insulin-like growth factor I isoform 1 preproprotein
    106 fibroblast growth factor 1 isoform 1 precursor
    107 fibroblast growth factor 2
    108 transforming growth factor beta-1 precursor
    109 transforming growth factor beta-2 isoform 1 precursor
    110 transforming growth factor beta-2 isoform 2 precursor
    111 actin, alpha skeletal muscle
    112 actin, aortic smooth muscle
    113 actin, cytoplasmic 1
    114 actin, alpha cardiac muscle 1 proprotein
    115 actin, cytoplasmic 2
    116 actin, gamma-enteric smooth muscle isoform 1 precursor
    117 actin, gamma-enteric smooth muscle isoform 2 precursor
    118 granulocyte colony-stimulating factor isoform a precursor
    119 granulocyte colony-stimulating factor isoform b precursor
    120 granulocyte colony-stimulating factor isoform c precursor
    121 granulocyte colony-stimulating factor isoform d precursor
    122 platelet-derived growth factor subunit A isoform 1
    preproprotein
    123 platelet-derived growth factor subunit A isoform 2
    preproprotein
    124 platelet-derived growth factor subunit B isoform 1
    preproprotein
    125 platelet-derived growth factor subunit B isoform 2
    preproprotein
    126 platelet-derived growth factor C precursor
    127 platelet-derived growth factor D isoform 1 precursor
    128 platelet-derived growth factor D isoform 2 precursor
    129 interstitial collagenase isoform 1 preproprotein
    130 interstitial collagenase isoform 2
    131 neutrophil collagenase preproprotein
    132 stromelysin-2 preproprotein
    133 macrophage metalloelastase preproprotein
    134 fibrillin-1 precursor
    135 fibrillin-2 precursor
    136 lysyl oxidase homolog 1 preproprotein
    137 lysyl oxidase homolog 2 precursor
    138 lysyl oxidase homolog 3 isoform 1 precursor
    139 lysyl oxidase homolog 3 isoform 2 precursor
    140 lysyl oxidase homolog 3 isoform 3
    141 lysyl oxidase homolog 4 precursor
    142 microfibrillar-associated protein 2 isoform a precursor
    143 microfibrillar-associated protein 2 isoform b precursor
    144 microfibrillar-associated protein 5 precursor
    145 disintegrin and metalloproteinase domain-containing
    protein 17 preprotein
    146 desmoglein-2 preproprotein
    147 DNA polymerase eta isoform 1
    148 DNA polymerase eta isoform 2
    149 DNA polymerase eta isoform 3
    150 ferrochelatase, mitochondrial isoform a precursor
    151 ferrochelatase, mitochondrial isoform b precursor
    152 filaggrin
    153 hyaluronan synthase 1 isoform 1
    154 hyaluronan synthase 1 isoform 2
    155 hyaluronan synthase 2
    156 hyaluronan synthase 3 isoform a
    157 hyaluronan synthase 3 isoform b
    158 proopiomelanocortin
    159 plakophilin-1 isoform 1a
    160 plakophilin-1 isoform 1b
    161 retinol dehydrogenase 10
    162 mitochondrial brown fat uncoupling protein 1
    163 tyrosinase precursor
    164 erythropoietin
    165 epoetin alfa
    166 darbepoetin alfa
    167 NOVEPOETIN
    168 NOVECRIT
  • This invention is further illustrated by the following non-limiting examples.
  • EXAMPLES Example 1 RNA Synthesis
  • RNA encoding green fluorescent protein (“GFP”), NOVEPOETIN (“EPO”), elastin (“ELN”), tyrosinase (“TYR”), melanocortin-1-receptor (“MC1R”), HAS1, HAS2, HAS3, COL3A1, COL7A1, COL1A1, COL1A2, hTERT, Holly GFP, Fresno RFP, Blitzen Blue, RIBOSLICE gene-editing proteins, TALENs, Cas9, Oct4, Sox2, Klf4, c-Myc-2 (T58A), Lin28, IL2, IL6, IL15, IL22, BMP2, BDNF, LIF, BMP6, IL15RA, FGF21, LIF, and PTH, and comprising various combinations of canonical and non-canonical nucleotides, was synthesized from DNA templates using the T7 High Yield RNA Synthesis Kit and the Vaccinia Capping System kit with mRNA Cap 2′-O-Methyltransferase (all from New England Biolabs, Inc.), according to the manufacturer's instructions and the present inventors' previously disclosed inventions (U.S. application Ser. No. 13/465,490 (now U.S. Pat. No. 8,497,124), International Application No. PCT/US12/67966, U.S. application Ser. No. 13/931,251, International Application No. PCT/US13/68118, and International Application No. PCT/US2015/013949, the contents of all of which are hereby incorporated by reference in their entirety) (Table 4). The RNA was then diluted with nuclease-free water to between 100 ng/μL and 2000 ng/μL. For certain experiments, an RNase inhibitor (Superase-In, Life Technologies Corporation) was added at a concentration of 1 μL/100 μg of RNA. RNA solutions were stored at room temperature, 4 C, −20 C or −80 C. For reprogramming experiments, RNA encoding Oct4, Sox2, Klf4, c-Myc-2 (T58A), and Lin28 was mixed at a molar ratio of 3:1:1:1:1.
  • TABLE 4
    RNA Synthesis
    Reaction
    Template Nucleotides Volume/μL ivT Yield/μg
    ELN A, 0.5 7dG, 0.4 5mU, 5mC 20 34.1
    ELN A, 0.5 7dG, 0.4 5mU, 5mC 20 67.6
    GFP A, 0.5 7dG, 0.4 5mU, 5mC 10 60.5
    GFP A, 0.5 7dG, 0.4 5mU, 5hmC 10 25.5
    GFP A, G, U, 5hmC 10 58.3
    GFP A, 0.5 7dG, U, 5hmC 10 47.3
    GFP A, 0.5 7dG, 0.4 5mU, 5cC 10 33.8
    GFP A, G, U, 5hmC 15 30.3
    GFP A, G, U, 5hmC 15 44.6
    GFP A, G, U, 5hmC 15 24.7
    TYR A, G, U, 5hmC 15 45.4
    MC1R A, G, U, 5hmC 15 47.5
    TYR A, G, U, C 20 67.0
    TYR A, G, psU, C 20 93.7
    TYR A, G, 0.4 5mU, C 20 85.7
    TYR A, G, U, 5mC 20 73.4
    TYR A, G, U, 5hmC 20 72.7
    TYR A, 0.5 7dG, U, C 20 62.7
    TYR A, G, psU, 5mC 20 116.3
    TYR A, G, psU, 5hmC 20 102.4
    TYR A, 0.5 7dG, psU, C 20 87.3
    TYR A, G, 0.4 5mU, 5mC 20 106.5
    TYR A, G, 0.4 5mU, 5hmC 20 85.0
    TYR A, 0.5 7dG, 0.4 5mU, C 20 70.9
    TYR A, 0.5 7dG, U, 5mC 20 88.5
    TYR A, 0.5 7dG, U, 5hmC 20 59.1
    TYR A, 0.5 7dG, psU, 5mC 20 7.8
    TYR A, 0.5 7dG, psU, 5hmC 20 98.0
    TYR A, 0.5 7dG, 0.4 5mU, 5mC 20 106.5
    TYR A, 0.5 7dG, 0.4 5mU, 5hmC 20 82.3
    HAS1 A, G, U, 5hmC 20 178.4
    HAS2 A, G, U, 5hmC 20 59.3
    HAS3 A, G, U, 5hmC 20 102.6
    TYR A, G, 0.4 5mU, 5hmC 100 377.3
    COL3A1 A, G, 0.4 5mU, 5hmC 20 108.3
    COL7A1 A, G, 0.4 5mU, 5hmC 20 94.6
    COL1A1 (20 μL) A, G, 0.4 5mU, 5hmC 20 114.0
    COL1A2 (10 μL) A, G, 0.4 5mU, 5hmC 10 31.3
    TYR A, G, 0.4 5mU, 5hmC 100 249.9
    GFP A, G, 0.4 5mU, 5hmC 100 264.0
    hTERT A, G, 0.4 5mU, 5hmC 100 349.2
    GFP A, G, U, 5hC 20 81.7
    GFP A, G, U, 0.5 5hC 20 65.4
    GFP A, sG, U, C 20 34.7
    GFP A, 0.5 sG, U, C 20 47.5
    GFP A, G, 5hmU, C 20 22.1
    GFP A, G, 0.5 5hmU, C 20 28.4
    GFP A, G, 5cU, C 20 24.4
    GFP A, G, 0.5 5cU, C 20 28.4
    GFP A, G, 5moU, C 20 39.2
    GFP A, G, 0.5 5moU, C 20 34.2
    GFP A, G, U, C 20 42.0
    GFP A, G, 5moU, C 20 53.8
    GFP A, G, 5moU, 5hmC 20 101.5
    GFP A, G, 0.4 5mU, 0.6 5moU, C 20 98.6
    GFP A, G, 0.4 5mU, C 20 99.6
    GFP A, G, U, 5mC 20 106.1
    GFP A, G, U, C 20 85.7
    GFP A, G, 5moU, C 100 398.4
    hTERT A, G, 5moU, C 20 82.6
    COL7A1 A, G, 5moU, C 20 34.9
    COL7A1 A, G, 5moU, C 100 342.0
    Holly GFP A, G, 5moU, C 20 36.7
    Fresno RFP A, G, 5moU, C 20 72.0
    Blitzen Blue A, G, 5moU, C 20 30.3
    hTERT A, G, 5moU, C 20 49.6
    Cas9 A, G, 5moU, C 20 31.6
    EPO A, G, U, C 20 101.0
    EPO A, G, 5moU, C 20 52.9
    EPO A, G, psU, C 20 106.0
    COL7A1 A, G, 5moU, C 20 80.2
    Oct4 (SEQ ID NO: 8) A, G, 5moU, C 300 1925.5
    Sox2 (SEQ ID NO: 9) A, G, 5moU, C 100 641.8
    Klf4 (SEQ ID NO: 10) A, G, 5moU, C 100 739.0
    c-Myc-2 (T58A) A, G, 5moU, C 100 574.0
    Lin28 A, G, 5moU, C 100 556.0
    IL2 A, G, 5moU, C 20 62.4
    IL6 A, G, 5moU, C 20 22.2
    IL15 A, G, 5moU, C 20 50.4
    IL22 A, G, 5moU, C 20 63.6
    BMP2 A, G, 5moU, C 20 83.2
    BDNF A, G, 5moU, C 20 45.0
    LIF A, G, 5moU, C 20 54.0
    BMP6 A, G, 5moU, C 20 92.2
    IL15RA A, G, 5moU, C 20 91.4
    FGF21 A, G, 5moU, C 20 79.2
    GFP A, G, 5moU, C 40 181.0
    IL2 A, G, 5moU, C 30 99.4
    IL6 A, G, 5moU, C 30 31.2
    IL15 A, G, 5moU, C 30 89.8
    IL22 A, G, 5moU, C 30 104.0
    BDNF A, G, 5moU, C 30 95.9
    BMP2 A, G, 5moU, C 30 112.0
    LIF A, G, 5moU, C 30 116.0
    PTH A, G, 5moU, C 30 88.4
    EPO A, G, 5moU, C 30 83.3
  • “A” refers to adenosine-5′-triphosphate, “G” refers to guanosine-5′-triphosphate, “U” refers to uridine-5′-triphosphate, “C” refers to cytidine-5′-triphosphate, “7dG” refers to 7-deazaguanosine-5′-triphosphate, “sG” refers to thienoguanosine-5′-triphosphate, “5mC” refers to 5-methylcytidine-5′-triphosphate, “5hmC” refers to 5-hydroxymethylcytidine-5′-triphosphate, “5cC” refers to 5-carboxycytidine-5′-triphosphate, “5fC” refers to 5-formylcytidine-5′-triphosphate, “5hC” refers to 5-hydroxycytidine-5′-triphosphate, “psU” refers to 5-pseudouridine-5′-triphosphate, “5mU” refers to 5-methyluridine-5′-triphosphate, “5hmU” refers to 5-hydroxymethyluridine-5′-triphosphate, “5cU” refers to 5-carboxyuridine-5′-triphosphate, and “5moU” refers to 5-methoxyuridine-5′-triphosphate.
  • Example 2 Preparation of RNA-Transfection-Reagent Complexes
  • For each microgram of RNA, 1 μg RNA and 1 μL transfection reagent (LIPOFECTAMINE 3000, Life Technologies Corporation) were first diluted separately in complexation medium (Opti-MEM, Life Technologies Corporation or DMEM/F12+10 μg/mL insulin+5.5 μg/mL transferrin+6.7 ng/mL sodium selenite+2 μg/mL ethanolamine) to a total volume of between 5 μL and 100 μL each. Diluted RNA and transfection reagent were then mixed and incubated for 10 min at room temperature, according to the transfection reagent-manufacturer's instructions.
  • Example 3 Transfection of Cells with Synthetic RNA
  • Complexes were prepared according to Example 2, and were then added directly to cells in culture. For transfection in 6-well plates, between 10 μL and 250 μL of complexes were added to each well of the 6-well plate, which already contained 2 mL of transfection medium per well. Plates were shaken gently to distribute the complexes throughout the well. Cells were incubated with complexes for 4 hours to overnight, before replacing the medium with fresh transfection medium (2 mL/well). Alternatively, the medium was not replaced. Volumes were scaled for transfection in 24-well and 96-well plates.
  • Example 4 Toxicity of and Protein Translation from Synthetic RNA Containing Non-Canonical Nucleotides
  • Primary human fibroblasts were transfected according to Example 2, using RNA synthesized according to Example 1. Cells were fixed and stained 20-24 h after transfection using an antibody against Oct4. The relative toxicity of the RNA was determined by assessing cell density at the time of fixation.
  • Example 5 Delivery of Synthetic RNA to the Skin
  • The complexation reaction shown in Table 5 was prepared using RNA encoding green fluorescent protein (GFP) or collagen, type VII, alphaI (COL7), synthesized according to Example 1. The concentration of the RNA stock solution was 500 μg/mL.
  • TABLE 5
    RNA Complexation Reaction
    Volume
    RNA solution tube
    GFP or COL7 RNA  8 μL
    FactorPlex ™ complexation buffer 42 μL
    Transfection reagent solution tube
    LIPOFECTAMINE 3000 (LIFE TECHNOLOGIES)  4 μL
    FactorPlex ™ complexation buffer 46 μL
  • Each tube was mixed by pipetting, and the transfection reagent solution tube was incubated for 30 s at room temperature. The transfection reagent solution was then transferred to the RNA solution, and the contents were mixed by rapidly pipetting up and down 10 times. Following a 10 min incubation, dilutions were prepared according to
  • TABLE 6
    Injection Solutions
    Complexation FactorPlex ™ RNA
    Site RNA Volume Volume amount
    1 GFP 7.5 μL  22.5 μL   0.3 μg
    2 GFP 15 μL 15 μL  0.6 μg
    3 GFP 30 μL 0 μL 1.2 μg
    4 COL7 30 μL 0 μL 1.2 μg
  • For each injection, the corresponding solution was drawn into a 3 cc insulin syringe with an 8 mm, 31 gauge needle (Becton, Dickinson and Company, Part Number: 328291) and air bubbles were removed. A clear field was selected on the left forearm of a healthy 33 year-old male human subject, and was disinfected with 70% isopropanol and allowed to dry. The needle was positioned at an angle of approximately 10° to the anterior (palmar) forearm with bevel facing up, and was inserted until the bevel was just covered. 30 μL of the RNA solution was injected intradermally over the course of approximately 10 sec. A distinct wheal appeared during the injection process. The needle was withdrawn, the wheal remained for approximately 1 minute, and no fluid escaped from the injection site. A total of 4 injections were performed according to Table 6, and all of the injections were performed between 11 and 28 minutes following the preparation of the RNA complexation reaction. No swelling, redness, or soreness occurred as a result of the injections. A small amount of bleeding occurred when the needle was removed from sites 2 and 4, resulting in the appearance of a small red spot at these sites.
  • The injection sites were imaged according to the schedule of Table 7, and every 24 hours thereafter for 6 days. Fluorescence images were acquired using an inverted microscope (Nikon Eclipse TS100) equipped with an EXFO X-Cite™ 120 fluorescence illumination system and the filter sets shown in Table 7. Fluorescence images were captured using a Sony NEX-7 digital camera (FIGS. 9-12).
  • TABLE 7
    Measurement Parameters
    Site Time Image Type Exposure Time
    All  0 h Brightfield Automatic
    1  0 h FITC 1/10 s
    2  0 h FITC 1/10 s
    3  0 h FITC 1/10 s
    4  0 h FITC 1/10 s
    1 12 h FITC 1/10 s
    2 12 h FITC 1/10 s
    3 12 h FITC 1/10 s
    4 12 h FITC 1/10 s
    1 12 h FITC 1/20 s
    2 12 h FITC 1/20 s
    3 12 h FITC 1/20 s
    4 12 h FITC 1/20 s
    All 24 h Brightfield Automatic
    1 24 h FITC 1/20 s
    2 24 h FITC 1/20 s
    3 24 h FITC 1/20 s
    4 24 h FITC 1/20 s
    1 24 h Cy3.5 s
    2 24 h Cy3.5 s
    3 24 h Cy3.5 s
    4 24 h Cy3.5 s
    1 24 h Cy3 s
    2 24 h Cy3 s
    3 24 h Cy3 s
    4 24 h Cy3 s
    1 36 h FITC 1/20 s
    2 36 h FITC 1/20 s
    3 36 h FITC 1/20 s
    4 36 h FITC 1/20 s
    1 48 h FITC 1/20 s
    2 48 h FITC 1/20 s
    3 48 h FITC 1/20 s
    4 48 h FITC 1/20 s
  • An independent experiment was carried out using the 1.2 μg dose of GFP RNA, with similar results (FIG. 12).
  • TABLE 8
    Filter Sets
    Image Type Filter Set
    Cy3 Chroma SP102V2
    Cy3.5 Chroma SP103V2
    FITC Chroma SP101
  • Example 6 Transfection of Human Keratinocytes with RNA Encoding NOVEPOETIN
  • RNA encoding NOVEPOETIN was synthesized according to Example 1 with three nucleotide combinations: 1) U, G, U, C, 2) U, G, 5moU, C, and 3) U, G, psU, C. Sub-confluent layers of primary human keratinocytes cultured in EpiLife medium were transfected in wells of a 6 well plate according to Example 3 with 1 μg of RNA per well. 12, 24, 36 and 48 h following transfection, 0.5 mL of medium was removed and 0.5 mL of fresh EpiLife medium was added to the plate. After the final medium sampling, the cells were harvested by trypsinization, and total RNA was isolated using the RNeasy mini kit (Qiagen). Genomic DNA was digested using DNase I and RNA was purified. Expression of interferon-β and GAPDH was measured by RT-PCR (FIG. 5).
  • Example 7 Transfection of Human Cells with RNA Encoding hTERT
  • RNA encoding human telomerase reverse transcriptase (hTERT) was synthesized according to Example 1 with the following nucleotides: U, G, 5moU, C. A sub-confluent layer of primary human dermal fibroblasts cultured in DMEM+10% FBS were transfected in wells of a 24 well plate according to Example 3 with 0.25 μg of RNA per well. 12 h after transfection, cells were fixed and stained using a 1:50 dilution of rabbit anti-hTERT antibody (Millipore, Part Number: MABE14) (FIG. 16).
  • Example 8 High-Efficiency Gene Editing by Repeated Transfection with RIBOSLICE
  • Primary human fibroblasts were plated in 6-well plates coated with recombinant human fibronectin and recombinant human vitronectin (each diluted in DMEM/F12 to a concentration of 1 μg/mL, 1 mL/well, and incubated at room temperature for 1 h) at a density of 10,000 cells/well in transfection medium. The following day, the cells were transfected as in Example 2 with RNA synthesized according to Example 1. The following day cells in one of the wells were transfected a second time. Two days after the second transfection, the efficiency of gene editing was measured using a mutation-specific nuclease assay.
  • Example 9 Transfection of Cells with Synthetic RNA Containing Non-Canonical Nucleotides and DNA Encoding a Repair Template
  • For transfection in 6-well plates, 1 μg RNA encoding gene-editing proteins targeting exon 16 of the human APP gene, 1 μg single-stranded repair template DNA containing a PstI restriction site that was not present in the target cells, and 6 μL transfection reagent (LIPOFECTAMINE RNAiMAX, Life Technologies Corporation) were first diluted separately in complexation medium (Opti-MEM, Life Technologies Corporation) to a total volume of 120 μL. Diluted RNA, repair template, and transfection reagent were then mixed and incubated for 15 min at room temperature, according to the transfection reagent-manufacturer's instructions. Complexes were added to cells in culture. Approximately 120 μL of complexes were added to each well of a 6-well plate, which already contained 2 mL of transfection medium per well. Plates were shaken gently to distribute the complexes throughout the well. Cells were incubated with complexes for 4 hours to overnight, before replacing the medium with fresh transfection medium (2 mL/well). The next day, the medium was changed to DMEM+10% FBS. Two days after transfection, genomic DNA was isolated and purified. A region within the APP gene was amplified by PCR, and the amplified product was digested with PstI and analyzed by gel electrophoresis.
  • Example 10 In Vivo RIBOSLICE Safety Study
  • 40 female NCr nu/nu mice were injected subcutaneously with 5×106 MDA-MB-231 tumor cells in 50% Matrigel (BD Biosciences). Cell injection volume was 0.2 mL/mouse. The age of the mice at the start of the study was 8 to 12 weeks. A pair match was conducted, and animals were divided into 4 groups of 10 animals each when the tumors reached an average size of 100-150 mm3, and treatment was begun. Body weight was measured every day for the first 5 days, and then biweekly to the end of the study. Treatment consisted of RIBOSLICE BIRC5-1.2 complexed with a vehicle (LIPOFECTAMINE 2000, Life Technologies Corporation). To prepare the dosing solution for each group, 308 μL of complexation buffer (Opti-MEM, Life Technologies Corporation) was pipetted into each of two sterile, RNase-free 1.5 mL tubes. 22 μL of RIBOSLICE BIRC5-1.2 (500 ng/μL) was added to one of the two tubes, and the contents of the tube were mixed by pipetting. 22 μL of vehicle was added to the second tube. The contents of the second tube were mixed, and then transferred to the first tube, and mixed with the contents of the first tube by pipetting to form complexes. Complexes were incubated at room temperature for 10 min. During the incubation, syringes were loaded. Animals were injected either intravenously or intratumorally with a total dose of 1 μg RNA/animal in 60 μL total volume/animal. A total of 5 treatments were given, with injections performed every other day. Doses were not adjusted for body weight. Animals were followed for 17 days. No significant reduction in mean body weight was observed, demonstrating the in vivo safety of RIBOSLICE gene-editing RNA.
  • Example 11 Screening of Reagents for Delivery of Nucleic Acids to Cells
  • Delivery reagents including polyethyleneimine (PEI), various commercial lipid-based transfection reagents, a peptide-based transfection reagent (N-TER, Sigma-Aldrich Co. LLC.), and several lipid-based and sterol-based delivery reagents were screened for transfection efficiency and toxicity in vitro. Delivery reagents were complexed with RIBOSLICE BIRC5-1.2, and complexes were delivered to HeLa cells in culture. Toxicity was assessed by analyzing cell density 24 h after transfection. Transfection efficiency was assessed by analyzing morphological changes. The tested reagents exhibited a wide range of toxicities and transfection efficiencies. Reagents containing a higher proportion of ester bonds exhibited lower toxicities than reagents containing a lower proportion of ester bonds or no ester bonds.
  • Example 12 High-Concentration Liposomal RIBOSLICE
  • High-Concentration Liposomal RIBOSLICE was prepared by mixing 1 μg RNA at 500 ng/μL with 3 μL of complexation medium (Opti-MEM, Life Technologies Corporation), and 2.5 μL of transfection reagent (LIPOFECTAMINE 2000, Life Technologies Corporation) per μg of RNA with 2.5 μL of complexation medium. Diluted RNA and transfection reagent were then mixed and incubated for 10 min at room temperature to form High-Concentration Liposomal RIBOSLICE. Alternatively, a transfection reagent containing DOSPA or DOSPER is used.
  • Example 13 In Vivo RIBOSLICE Efficacy Study—Subcutaneous Glioma Model
  • 40 female NCr nu/nu mice were injected subcutaneously with 1×107 U-251 tumor cells. Cell injection volume was 0.2 mL/mouse. The age of the mice at the start of the study was 8 to 12 weeks. A pair match was conducted, and animals were divided into 4 groups of 10 animals each when the tumors reached an average size of 35-50 mm3, and treatment was begun. Body weight was measured every day for the first 5 days, and then biweekly to the end of the study. Caliper measurements were made biweekly, and tumor size was calculated. Treatment consisted of RIBOSLICE BIRC5-2.1 complexed with a vehicle (LIPOFECTAMINE 2000, Life Technologies Corporation). To prepare the dosing solution, 294 μL of complexation buffer (Opti-MEM, Life Technologies Corporation) was pipetted into a tube containing 196 μL of RIBOSLICE BIRC5-1.2 (500 ng/μL), and the contents of the tube were mixed by pipetting. 245 μL of complexation buffer was pipetted into a tube containing 245 μL of vehicle. The contents of the second tube were mixed, and then transferred to the first tube, and mixed with the contents of the first tube by pipetting to form complexes. Complexes were incubated at room temperature for 10 min. During the incubation, syringes were loaded. Animals were injected intratumorally with a total dose of either 2 μg or 5 μg RNA/animal in either 20 μL or 50 μL total volume/animal. A total of 5 treatments were given, with injections performed every other day. Doses were not adjusted for body weight. Animals were followed for 25 days.
  • Example 14 Liposome Formulation and Nucleic-Acid Encapsulation
  • Liposomes are prepared using the following formulation: 3.2 mg/mL N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-DSPE), 9.6 mg/mL fully hydrogenated phosphatidylcholine, 3.2 mg/mL cholesterol, 2 mg/mL ammonium sulfate, and histidine as a buffer. pH is controlled using sodium hydroxide and isotonicity is maintained using sucrose. To form liposomes, lipids are mixed in an organic solvent, dried, hydrated with agitation, and sized by extrusion through a polycarbonate filter with a mean pore size of 800 nm. Nucleic acids are encapsulated by combining 10 μg of the liposome formulation per 1 μg of nucleic acid and incubating at room temperature for 5 minutes.
  • Example 15 Folate-Targeted Liposome Formulation
  • Liposomes are prepared using the following formulation: 3.2 mg/mL N-(carbonyl-ethoxypolyethylene glycol 2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-DSPE), 9.6 mg/mL fully hydrogenated phosphatidylcholine, 3.2 mg/mL cholesterol, 2 mg/mL ammonium sulfate, and histidine as a buffer, with 0.27 mg/mL 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[folate(polyethylene glycol)-5000] (FA-MPEG5000-DSPE) added to the lipid mixture. pH is controlled using sodium hydroxide and isotonicity is maintained using sucrose. To form liposomes, lipids are mixed in an organic solvent, dried, hydrated with agitation, and sized by extrusion through a polycarbonate filter with a mean pore size of 800 nm. Nucleic acids are encapsulated by combining 10 μg of the liposome formulation per 1 μg of nucleic acid and incubating at room temperature for 5 minutes.
  • Example 16 Therapy Comprising Liposomal Protein-Encoding RNA
  • Liposomes encapsulating synthetic RNA encoding a therapeutic protein, synthesized according to Example 1, are prepared according to Example 14 or Example 15. The liposomes are administered by injection or intravenous infusion.
  • Example 17 Generation of Elastin ivT-RNA Template
  • Total RNA was extracted from neonatal human dermal fibroblasts using the RNeasy mini kit (QIAGEN GmbH), according to the manufacturer's instructions. cDNA encoding human elastin was prepared using MonsterScript™ Reverse Transcriptase (Epicentre Biotechnologies) and the primer: AAAAAAACCGGT TCATTTTCTCTTCCGGCCAC (SEQ ID NO:483). An in vitro transcription (ivT) template was prepared from the cDNA by PCR amplification of the elastin coding sequence (CDS) using the primers: F: AAAAAAGCTAGCATGGCGGGTCTGACG (SEQ ID NO:484), and R: AAAAAAACCGGTTCATTTTCTCTTCCGGCCAC (SEQ ID NO:485). The PCR product was then purified using agarose gel electrophoresis and the QIAquick Gel Extraction Kit (QIAGEN GmbH) and was cloned into a vector containing the human beta globin (HBB) 5′ and 3′ untranslated regions and a strong Kozak sequence. The vector was amplified, purified, and linearized prior to RNA synthesis.
  • Example 18 Generation of Tyrosinase ivT-RNA Template
  • Total RNA was extracted from human epidermal melanocytes using the RNeasy mini kit (QIAGEN GmbH), according to the manufacturer's instructions. cDNA encoding human tyrosinase was prepared using MonsterScript™ Reverse Transcriptase (Epicentre Biotechnologies). An in vitro transcription (ivT) template was prepared from the cDNA by PCR amplification of the tyrosinase coding sequence (CDS). The PCR product was then purified using agarose gel electrophoresis and the QIAquick Gel Extraction Kit (QIAGEN GmbH) and was cloned into a vector containing the human beta globin (HBB) 5′ and 3′ untranslated regions and a strong Kozak sequence. The vector was amplified, purified, and linearized prior to RNA synthesis.
  • Example 19 Synthesis of Tyrosinase RNA
  • RNA encoding human tyrosinase was synthesized according to Example 1, using the DNA template of Example 18 and the T7 High Yield RNA Synthesis Kit (New England Biolabs, Inc.), according to the manufacturer's instructions (Table 4). Samples of the RNA were analyzed by agarose gel electrophoresis to assess the quality of the RNA. The RNA was then diluted to 1 μg/μL. The RNA solution was stored at 4 C.
  • Example 20 Increasing Melanin Production in Skin by Transdermal Injection Via Syringe of RNA Encoding Tyrosinase
  • The RNA of Example 19 was loaded into a syringe and delivered to the dermis of the ventral forearm of a healthy 33 year-old male patient over the course of approximately 30 seconds.
  • Example 21 Increasing Melanin Production in Skin by Combined Delivery of RNA Encoding Tyrosinase and Electroporation
  • The area of skin treated in Example 20 was exposed to electrical pulses of between 10V and 155V and between approximately 10 milliseconds and approximately 1 second using a two-electrode array electrically connected to a capacitor. The patient reported a tingling sensation at all voltages and penetration depths. The treated area became darker after 24-48 hours. The experiment was repeated several times, with similar results.
  • Example 22 Increasing Melanin Production in Skin by Topical or Intradermal Application of RNA Encoding Tyrosinase
  • The RNA of Example 19 or the liposomes of Example 16 are applied directly to the skin, with or without disruption of the stratum corneum or injected intradermally using a dose of one microgram or less per square centimeter. Optionally, an electric field is applied as in Example 21 or using a surface-contact patch to enhance delivery of the RNA.
  • Example 23 Increasing Elastin Production in Skin by Transdermal Delivery of RNA Encoding Elastin
  • RNA encoding elastin was prepared according to Example 1. The RNA is delivered as in Example 20, 21, or 22.
  • Example 24 Increasing Collagen Production in Skin by Transdermal Delivery of RNA Encoding Collagen
  • RNA encoding collagen was prepared according to Example 1. The RNA is delivered as in Example 20, 21, or 22.
  • Example 25 Anemia Therapy Comprising Delivery of RNA Encoding NOVEPOETIN
  • RNA encoding NOVEPOETIN was prepared according to Example 1. The RNA is delivered as in Example 20, 21, or 22.
  • Example 26 Increasing Production of Actin in Skeletal Muscle by Intramuscular Delivery of RNA Encoding Actin
  • RNA encoding actin is prepared according to Example 1. The RNA is delivered to the patient via intramuscular injection with or without the use of an electric field as in Example 20, 21, or 22.
  • Example 27 Wound Healing Treatment
  • RNA encoding basic fibroblast growth factor is prepared according to Example 1. The RNA is delivered as in Example 20, 21, or 22.
  • Example 28 Anti-Scarring Treatment
  • RNA encoding collagenase is prepared according to Example 1. The RNA is delivered as in Example 20, 21, or 22.
  • Example 29 Production of Botulinum Toxin
  • RNA encoding botulinum toxin is prepared according to Example 1. The RNA is delivered as in Example 20, 21, or 22.
  • Example 30 Increasing Collagen Production in Skin Cells by Transfection with RNA Encoding Collagen I
  • RNA comprising the coding sequence of the human COL1A1 gene was synthesized according to Example 1. Primary human dermal fibroblasts were plated in wells of a 24-well plate, and were transfected according to Example 2. Between 24 and 72 hours after transfection, the cells were fixed and stained using an antibody targeting collagen I. Many extracellular deposits of collagen were visible in the transfected wells.
  • Example 31 Increasing Collagen Production in Skin Cells by Transfection with RNA Encoding Collagen VII
  • RNA comprising the coding sequence of the human COL7 gene was synthesized according to Example 1. Primary human dermal fibroblasts were plated in wells of a 24-well plate, and were transfected according to Example 2. Between 24 and 72 hours after transfection, the cells were fixed and stained using an antibody targeting collagen VII. Transfected cells exhibited high levels of collagen VII, compared to an un-transfected control.
  • Example 32 Increasing Collagen Production in Skin by Transdermal Injection Via Syringe of RNA Encoding Collagen I or Collagen VII
  • RNA comprising the coding sequence of the human COL1A1 gene or the human COL7 gene was synthesized according to Example 1. The RNA is loaded into a syringe and delivered to the dermis of a patient over the course of approximately 30 seconds or as in Example 20, 21, or 22.
  • Example 33 Increasing Collagen Production in Skin by Combined Delivery of RNA Encoding Collagen I or Collagen VII and Electroporation
  • The area of skin treated in Example 32 is exposed to electrical pulses of between 10V and 155V and between approximately 50 microseconds and approximately 1 second using a multi-electrode array electrically connected to a power source.
  • Example 34 Storage and Stability of Synthetic RNA Complexes
  • A complexation reaction using RNA encoding GFP was prepared according to Example 5. Following the 10 min incubation, the complexation reaction was divided into three equal parts, one of which was diluted 1:10 in FactorPlex™ complexation medium, one of which was diluted 1:10 in sterile, nuclease-free water, and one of which was left undiluted. Each of the three parts was then further divided into four equal parts, one of which was applied to primary human dermal fibroblasts according to Example 3, one of which was left at room temperature for six hours before applying to primary human dermal fibroblasts according to Example 3, one of which was placed at 4° C. for six hours before applying to primary human dermal fibroblasts according to Example 3, and one of which was snap frozen in liquid nitrogen and placed at −80° C. for six hours before applying to primary human dermal fibroblasts according to Example 3. The cells were imaged using a fluorescence microscope approximately 24 hours after the first transfection (FIG. 17). All wells contained GFP-positive cells, demonstrating that the synthetic RNA complexes were stable and maintained activity in all of the storage conditions tested.
  • Example 35: In Vivo Analysis of NOVECRIT
  • RNA encoding NOVEPOETIN was synthesized according to Example 1 with the nucleotide combination A, G, 5moU, C. In this Example, NOVECRIT was formulated with a lipid delivery vehicle, specifically LIPOFECTAMINE 3000. In this Example, NOVECRIT encoded NOVEPOETIN, a novel, high-stability erythropoiesis-stimulating agent.
  • A 15-day maximum tolerated dose (MTD) study was performed to evaluate safety (rat, n=62). In vivo toxicology and biodistribution, as well as pharmacodynamics and dose response and therapeutic effect, specifically on erythropoiesis, were evaluated. Furthermore, the immune response was monitored by analysis of cytokines in treated animals.
  • Sixty-nine, naïve, 8 week old male Sprague-Dawley rats (Rattus norvegicus) weighing 253 to 274 grams at receipt were used (HARLAN LABORATORIES). Animals were acclimated to the study room for at least seven days prior to dosing. On Day −2 all animals were shaved in the dorsal lumbar area and four intradermal sites (upper left, upper right, lower right and lower left) were designated on the dorsal back using a permanent marker. Sixty-four animals were randomly assigned to four treatment groups, with the remaining five animals serving as spares. Two animals were dropped from the study because of incomplete dosing.
  • The following study design was used (total dose volumes (μL) were constant):
  • Dose Dose
    Level Dose Conc. Vol.
    Group (μg) Route (μg/mL) (μL) Number of Males
    1 0 Intra- 0 50 4a + 2b + 2h
    dermal
    2 0.25 Intra- 5.0 50 4a + 2b + 2c + 2d +
    dermal 2e + 2f + 2g + 2h
    3 1.0 Intra- 20 50 4a + 2b + 2c + 2d +
    dermal 2e + 2f + 2g + 2h
    4 4 × 1.0 μg Intra- 20 50 4a + 2b + 3c + 3d +
    (4.0 μg total) dermal 2e + 2f + 2g + 2h
    ID: aToxicology animals necropsy on Day 15, TK animals with terminal tissue collections on Days 6b and 24c hours postdose, and on Days 3d, 4e, 6f, 8g, and 15h
  • Blood was collected from the vena cava from anesthetized animals prior to necropsy or terminal tissue collection. Whenever possible, blood was collected via a single draw and then divided appropriately. Blood specimens for toxicokinetic analysis were collected from two animals per time point for Group 1 at 6 hours postdose and on Day 15. For Groups 2-4, blood specimens were collected at 6 and 24 hours postdose, and on Days 3, 4, 6, 8, and 15.
  • Blood specimens for hematology assessment were collected from all toxicology animals on Day 15 following an overnight fast, and all TK animals scheduled for terminal tissue collections at 6 and 24 hours postdose and on Days 8 and 15. Whole blood (1.3 mL) was deposited into K2EDTA tubes and analyzed using an Advia 120 automated analyzer.
  • Blood specimens for coagulation assessment were collected from all toxicology animals on Day 15 following an overnight fast. Coagulation specimens (1.8 mL) were collected into 3.2% sodium citrate tubes, processed according to standard procedures and analyzed using a STA Compact automated analyzer.
  • Blood specimens for serum chemistry assessments were collected from all toxicology animals on Day 15 following an overnight fast. Serum chemistry specimens (1 mL) were collected into serum separator tubes, processed according to standard procedures and analyzed using an AU680 analyzer.
  • Blood specimens for cytokine analysis were collected from animals scheduled for terminal tissue collection at 5 and 24 hours postdose and Day 8. Cytokine specimens (1 mL) were collected into K2EDTA tubes and processed to plasma according to standard procedures. TNFα, IL-6, and IFNα cytokine levels in plasma samples were studied. The study measured the production of TNFα, IL-6, and IFNα cytokines in plasma samples following dosing with the test article on Day 1 (6 hours post-dose), Day 2 (24 hours post-dose) and Day 8.
  • Study Outline for Cytokine Analysis Samples:
  • Dose
    NOVECRIT Concen- Dose Number
    Dose Level tration Volume of Rats
    Group (μg) (μg/mL) (μL) (Males) Endpoints
    1 0 0 50 2b Plasma TNFα,
    2 0.25 5.0 50 2b + 2c + 2g IL-6, and
    3 1.0 20 50 2b + 2c + 2g IFNα cytokine
    4 4 × 1.0 μg 20 50 2b + 2c + 2g analysis
    (4.0 μg total)
    b = plasma samples collected 6 hours post-dose, c = plasma samples collected 24 hours post-dose, g = plasma samples collected 8 days post-dose.
  • For TNFα analysis, plasma samples were analyzed using a commercially-available assay kit from R&D SYSTEMS (cat. no. RTA00). The rat TNFα ELISA is a solid phase enzyme-linked immunosorbent assay. The ELISA kit employs a TNFα-specific anti-rat monoclonal antibody for solid phase immobilization (pre-coated on the microtiter plate), and HRP conjugated to an anti-rat TNFα polyclonal antibody for detection. The reference standard provided with the kit is a recombinant rat TNFα. For each assay, plasma samples and standards were diluted with the diluent provided in each respective kit. For the TNFα ELISA, plasma samples were diluted 1:2.
  • The standard curve consisted of six (6) serial 2-fold concentrations ranging from 800 to 12.5 pg/mL. Controls were reconstituted with diluent, and blanks contained diluent only.
  • Following sample, standard, control, or diluent addition (50 μL per well, each in duplicate), plates were incubated for 2 hours at room temperature. Following a wash step to remove unbound substances, HRP conjugate was added to each well (100 μL/well), and plates were incubated for 2 hours at room temperature. Following a second wash step, 100 μL/well TMB substrate was added to the plate. The plate was incubated for 30 minutes at room temperature, protected from light, to allow the color reaction to develop. The reaction was stopped following the addition of HCl Stop Solution (100 μL/well). The optical density was read on a SpectraMax 340 (MOLECULAR DEVICES) plate reader at 450 nm, within 30 minutes following addition of Stop Solution. The intensity of the color measured was in proportion to the amount of rat TNFα bound in the initial step. A standard curve was generated for each assay plate, and test sample TNFα concentrations were determined by interpolation of absorbance A450 values from the standard curve and dilution factor. The assay range for the TNFα kit is 12.5-800 pg/mL, with a minimum detectable concentration of less than 10 pg/mL (with the 1:2 minimum required dilution for plasma samples).
  • For IL-6 analysis, plasma IL-6 samples were analyzed using a commercially-available assay kit from R&D SYSTEMS (cat. no. R6000B—IL-6). The rat IL-6 ELISA is a solid phase enzyme-linked immunosorbent assay. The ELISA kit employs anti-rat IL-6 monoclonal antibody for solid phase immobilization (pre-coated on the microtiter plate), and HRP conjugated to IL-6-specific anti-rat polyclonal antibody for detection. The reference standard provided with the kit is a recombinant rat IL-6. Plasma samples were used undiluted (as provided), and standards were diluted with the diluent provided in the kit. The standard curve consisted of six (6) serial 2-fold concentrations ranging from 4,000 to 62.5 pg/mL. Controls were reconstituted with diluent, and blanks contained diluent only. Following sample, standard, control, or diluent addition (50 μL per well, each in duplicate), plates were incubated for 2 hours at room temperature. Following a wash step to remove unbound substances, HRP conjugate was added to each well (100 μL/well), and plates were incubated for 2 hours at room temperature. Following a second wash step, 100 μL/well TMB substrate was added to the plate. The plate was incubated for 30 minutes at room temperature, protected from light, to allow the color reaction to develop. The reaction was stopped following the addition of HCl Stop Solution (100 μL/well). The optical density was read on a SpectraMax 340 (MOLECULAR DEVICES) plate reader at 450 nm, within 30 minutes following addition of Stop Solution. The intensity of the color measured was in proportion to the amount of rat IL-6 bound in the initial step. A standard curve was generated for each assay plate, and test sample IL-6 concentrations were determined by interpolation of absorbance A450 values from the standard curve and dilution factor. The assay range for the IL-6 kit is 62.5-4,000 pg/mL, with a minimum detectable concentration of less than 21 pg/mL.
  • For IFNα analysis, plasma IFNα samples were analyzed using a commercially-available assay kit from NOVATEINBIO (cat. no. BG-RAT11380). The ELISA is a solid phase enzyme-linked immunosorbent assay. The ELISA kit employs anti-rat IFNα monoclonal antibody for solid phase immobilization (pre-coated on the microtiter plate), and HRP conjugated antibody specific for IFNα for detection. Plasma samples were diluted 1:4, and the standards were used as provided in the kit. Standards consisted of six (6) serial 2-fold concentrations ranging from 100 to 3.1 pg/mL. Blanks contained diluent only. Following sample, standard, or diluent addition (50 μL per well, each in duplicate), HRP conjugate was added to each well (100 μL/well), and plates were incubated for 1 hours at 37° C. Following a wash step, 50 μL/well each of Chromogen Solution A and Chromogen Solution B were added to the plate. The plate was incubated for 15 minutes at 37° C., protected from light, to allow the color reaction to develop. The reaction was stopped following the addition of Stop Solution (50 μL/well). The optical density was read on a SpectraMax 340 (MOLECULAR DEVICES) plate reader at 450 nm. The intensity of the color measured was in proportion to the amount of rat IFNα bound in the initial step. A standard curve was generated for each assay plate, and test IFNα concentrations were determined by interpolation of absorbance A450 values from the standard curve and dilution factor. The assay range for the IFNα kit is 3.1-100 pg/mL, with a minimum detectable concentration of less than 1 pg/mL (4 pg/mL, with the 1:4 dilution required for plasma samples).
  • The results of this study were, inter alia, that NOVECRIT was not detected at the injection site 24 h after dosing (as assayed by RT-PCR). More specifically, NOVECRIT was not detected in any of the following samples (RT-PCR): serum (6 h, 24 h, 48 h and 72 h after dosing), liver (6 h and 24 h after dosing), and kidney (6 h and 24 h after dosing). Further, positive controls (spiked with NOVECRIT) yielded a robust signal in all tissues (as assayed RT-PCR). FIG. 18 depicts single administration of NOVECRIT induced a rapid increase and sustained level of NOVEPOIETIN in serum. The Y axis shows concentration of NOVEPOIETIN protein (mU/mL). This suggests, without wishing to be bound by theory, that the studied RNA therapeutic is able to provide therapeutically important pharmacodynamic properties. In fact, this PD behavior is vastly improved relative to wild type EPO (which is known in the art to have a half-life of about 4-12 hours).
  • Furthermore, serum spiked with NOVECRIT showed near-instant degradation of the RNA (as assayed by RT-PCR). Without wishing to be bound by theory, this data indicates that the present RNA therapeutics are safe and have little chance of toxicity limitations, for example, liver or kidney toxicities.
  • FIG. 19 depicts a single administration of NOVECRIT stimulated erythropoiesis, yielding elevated hematocrit for at least 14 days. The left panel shows % hematocrit on the Y axis, while the right panel shows % reticulocytes. Accordingly, the studied RNA therapeutic is fully functional.
  • With respect to cytokines, IL-6 and TNFα cytokine levels were below assay detection thresholds for all animals at each of the timepoints evaluated in this study. Low levels of IFN-□□ were only detectable in the Day 8 samples from animals in Groups 3 and 4. FIG. 20 depicts a table summarizing TNFα, IL-6, and IFNα cytokine levels in plasma samples collected from a maximum tolerated dose of NOVECRIT in male Sprague Dawley rats. Without wishing to be bound by theory, this data indicates that the present RNA therapeutics do not stimulate an unfavorable immunogenicity which has limited the therapeutic utility of certain RNA therapeutics.
  • Example 36: Gene Editing of the COL7A Gene
  • The present RNA-based gene editing approaches were applied to the COL7A1 gene. This gene is of interest because, inter alia, it is frequently involved in dystrophic epidermolysis bullosa. FIG. 21 depicts a SURVEYOR assay using the DNA of primary adult human dermal fibroblasts transfected with RNA TALENs targeting the sequence TGAGCAGAAGTGGCTCAGTG (SEQ ID NO:467) and TGGCTGTACAGCTACACCCC (SEQ ID NO:468), located within the COL7A1 gene. The bands present in the +RNA lane indicate editing of a region of the gene that is frequently involved in dystrophic epidermolysis bullosa. FIG. 22 depicts another SURVEYOR assay using the DNA of primary adult human dermal fibroblasts transfected with RNA TALENs, now targeting the sequence TTCCACTCCTGCAGGGCCCC (SEQ ID NO:469) and TCGCCCTTCAGCCCGCGTTC (SEQ ID NO:470), located within the COL7A1 gene. The bands present in the +RNA lane indicate editing of a region of the gene that is frequently involved in dystrophic epidermolysis bullosa. This data points to, among others, a gene editing approach to the treatment of certain genetic disorders such as dystrophic epidermolysis bullosa.
  • Example 37: Gene-Editing of the MYC Gene Using a Synthetic RNA with Non-Canonical Nucleotides
  • Experiments were conducted with in vitro transcribed synthetic RNA molecules containing non-canonical nucleotides and encoding gene-editing proteins. The immunogenicity and the gene-editing efficiency of in vitro transcribed synthetic RNA molecules having (1) only pseudouridine (psU) as a non-canonical nucleotide; (2) only 5-methylcytidine (5mC) as a non-canonical nucleotide; and (3) both of pseudouridine and 5-methylcytidine as non-canonical nucleotides was evaluated (as well as controls).
  • Specifically, mRNA containing the following nucleotide combinations: (i) A,G,U,C, (ii) A,G,psU,C, (iii) A,G,U,5mC, and (iv) A,G,psU,5mC, and encoding TALEN pairs targeting the following DNA sequences, which can be found within the MYC gene: TCGGCCGCCGCCAAGCTCGT (SEQ ID NO:474) and TGCGCGCAGCCTGGTAGGAG (SEQ ID NO:475), were synthesized according to the methods described herein. Human dermal fibroblasts (MA001SK) were plated in 6-well and 24-well tissue culture plates in DMEM with 10% FBS at 100,000 and 10,000 cells per well, respectively. The next day, the cells were transfected in the 6-well plate with 2 μg of RNA (1 μg for each component of the TALEN pair) and the cells were transfected in the 24-well plate with 0.2 μg of RNA (0.1 μg for each component of the TALEN pair) according to the methods described herein. 24 hours after transfection, the total RNA from the cells in the 24-well culture plate was isolated using an RNeasy Mini Kit (74106; QIAGEN), including isolating the total RNA from a sample of cells that had not been transfected with RNA (negative control; “Neg.” in FIG. 23). The genomic DNA was removed by a 15 minute digestion with DNase I (RNase-Free) (M0303L; NEW ENGLAND BIOLABS) and the reaction purified using an RNeasy Mini Kit. 1 μL of total RNA was used to assess gene expression by real-time RT-PCR using TAQMAN gene-expression assays (APPLIED BIOSYSTEMS) designed to detect expression of the immunogenicity markers TLR3, IFIT1, and IFIT2 (FIG. 23). The data were normalized to both the positive experimental control sample (“A,G,U,C”) and to a loading control (GAPDH). 48 hours after transfection, the genomic DNA was isolated from the cells in the 6-well culture plate using a DNeasy Blood and Tissue Kit (69506; QIAGEN), including from a sample of cells that had not been transfected with RNA (negative control, “Neg.” in FIG. 24). A 970 bp region of the MYC gene surrounding the predicted TALEN cut location was amplified using a 35 cycle 2-step PCR reaction containing the following primers: TAACTCAAGACTGCCTCCCGCTTT (SEQ ID NO: 476) and AGCCCAAGGTTTCAGAGGTGATGA (SEQ ID NO: 477). 160 ng was hybridized in 5 μL of amplified sequence from RNA-treated cells to 160 ng in 5 μL of amplified sequences from untreated MA001SK cells by mixing the two sequences with 0.5 μL of 1M KCl and 0.5 μL of 25 mM MgCl2 and running the following program in a thermocycler: 95° C. for 10 minutes; 95° C. to 85° C. at 0.625 C/s; 85° C. to 25° C. at 0.125 C/s. The SURVEYOR assay was performed by adding 0.5 μL of SURVEYOR nuclease and 0.5 μL of Enhancer from the SURVEYOR Mutation Detection Kit (7060201; INTEGRATED DNA TECHNOLOGIES) to the hybridized product, mixing, and incubating at 42° C. for 25 minutes. The protocol above was also used to process the positive control DNA sample provided with the SURVEYOR Mutation Detection Kit as a positive experimental control for the SURVEYOR Assay (“Assay Pos.” in FIG. 24). Samples were analyzed by agarose gel electrophoresis (FIG. 24). For each sample, gene-editing efficiency was calculated as a ratio of the intensity of the digested bands (indicated by “*” in FIG. 24) to that of the undigested band.
  • As shown in FIG. 23 below, the samples from cells transfected with the positive control RNA (A,G,U,C), and the samples from cells transfected with RNA containing either pseudouridine or 5-methylcytidine exhibited upregulation of all three of the immunogenicity markers TLR3, IFIT1, and IFIT2. The sample from cells transfected with RNA containing both pseudouridine and 5-methylcytidine exhibited negligible upregulation of the immunogenicity markers (less than 0.01-fold of the positive control), demonstrating that in vitro transcribed synthetic RNA with both pseudouridine and 5-methylcytidine and encoding a gene-editing protein can evade detection by the innate-immune system of mammalian cells.
  • Further, as shown in FIG. 24 below, the sample from cells transfected with RNA containing both pseudouridine and 5-methylcytidine exhibited highly efficient gene editing (41.7%), which was greater than the efficiency exhibited by samples from cells transfected with RNA containing pseudouridine alone (35.2%), demonstrating that in vitro transcribed synthetic RNA comprising both pseudouridine and 5-methylcytidine and encoding a gene-editing protein can both (i) gene-edit mammalian cells at high efficiency, and (ii) gene-edit mammalian cells at higher efficiency than in vitro transcribed synthetic RNA comprising pseudouridine and not comprising 5-methylcytidine.
  • Example 38: COL7A1 Gene Editing and Repair in Human Cells
  • RNA encoding gene editing proteins targeting the following sequences in the COL7A1 gene was synthesized according to Example 1: TGAGCAGAAGTGGCTCAGTG (SEQ ID NO:473) and TGGCTGTACAGCTACACCCC (SEQ ID NO:468) (see also table below).
  • RNA Synthesis
  • Reaction
    Template Nucleotides Volume/μL ivT Yield/μg
    COL7A1 TALEN 1L A, G, 5moU, C 20 120.528
    COL7A1 TALEN 1R A, G, 5moU, C 20 120.204
    COL7A1 TALEN 1L A, G, 5moU, C 15 81.94
    COL7A1 TALEN 1R A, G, 5moU, C 15 61.88
  • 50,000 primary human epidermal keratinocytes (HEKn, Gibco) were plated in wells of 6-well plates in EpiLife+Supplement S7. The next day, cells were transfected according to Example 3 with 1 μg of RNA encoding each component of the gene editing pair and 2 μg of a single-stranded DNA repair template having a length of 60, 70, 80, 90 or 100 nucleotides (“nt”). 48 hours after transfection, genomic DNA was purified. A segment of the COL7A1 gene was amplified using the primers GCATCTGCCCTGCGGGAGATC (SEQ ID NO:478) and CCACGTTCTCCTTTCTCTCCCCGTTC (SEQ ID NO:479), which produce a 535 bp amplicon. The efficiency of gene editing was assessed using T7 Endonuclease I (“T7E1”, New England Biolabs) according to the manufacturer's instructions. Bands of approximately 385 bp and 150 bp indicate successful gene editing. FIG. 25 and FIG. 29 show the result of digestion with T7EI, analyzed by agarose gel electrophoresis. FIG. 27 and FIG. 29 show the result of digestion with MluI-HF, analyzed by agarose gel electrophoresis. Because the repair template contains the sequence ACGCGT (SEQ ID NO:480), digestion of the amplified product with MluI-HF (New England Biolabs) produces bands of approximately 385 bp and 150 bp in the case of successful gene repair.
  • RNA encoding gene editing proteins targeting the following sequences in the COL7A1 gene was synthesized according to Example 1: TGAGCAGAAGTGGCTCAGTG (SEQ ID NO:473) and TGGCTGTACAGCTACACCCC (SEQ ID NO:468). 50,000 primary human epidermal keratinocytes (HEKn, Gibco) were plated in wells of 6-well plates in EpiLife+Supplement S7. The next day, cells were transfected according to Example 3 with 1 μg of RNA encoding each component of the gene editing pair and 1-4 μg of an 80 nucleotide single-stranded DNA repair template. 48 hours after transfection, genomic DNA was purified. A segment of the COL7A1 gene was amplified using the primers GCATCTGCCCTGCGGGAGATC (SEQ ID NO:481) and CCACGTTCTCCTTTCTCTCCCCGTTC (SEQ ID NO:482), which produce a 535 bp amplicon. The efficiency of gene editing was assessed using T7 Endonuclease I (“T7E1”, New England Biolabs) according to the manufacturer's instructions. Bands of approximately 385 bp and 150 bp indicate successful gene editing. FIG. 26 show the result of digestion with T7EI, analyzed by agarose gel electrophoresis. FIG. 28 show the result of digestion with MluI-HF, analyzed by agarose gel electrophoresis. Because the repair template contains the sequence ACGCGT (SEQ ID NO:480), digestion of the amplified product with MluI-HF (New England Biolabs) produces bands of approximately 385 bp and 150 bp in the case of successful gene repair.
  • Example 39: Expression of BMP7 Variants in Human Cells
  • RNA encoding wild type BMP7 and RNA encoding variants of BMP7 was synthesized according to Example 1 (see also table below).
  • RNA Synthesis
  • Reaction
    Template Nucleotides Volume/μL ivT Yield/μg
    BMP7 Wild Type A, G, 5moU, C 15 125.8
    BMP7 Variant A A, G, 5moU, C 15 120.36
    BMP7 Variant B A, G, 5moU, C 15 143.14
    BMP7 Variant C A, G, 5moU, C 15 106.42
  • 50,000 primary human dermal fibroblasts (MA001SK, Factor Bioscience) or 100,000 primary human epidermal keratinocytes (HEKn, Gibco) were plated in DMEM+10% FBS or EpiLife+Supplement S7, respectively. Cells were transfected according to Example 3 with 1 μg of RNA encoding wild type BMP7 or a variant thereof. 24 hours after transfection, the medium was sampled and secreted BMP7 levels were measured with a human BMP7 ELISA kit (ab99985, Abcam) using medium diluted 10-fold according to the manufacturer's instructions. Secreted BMP7 levels were determined by measuring 450 nm absorbance using a microplate reader (EMax Plus, Molecular Devices). Secreted BMP7 levels are shown in FIG. 30.
  • Example 40: Expression of Parathyroid Hormone (PTH) in Human Cells
  • RNA encoding PTH was synthesized according to Example 1 (see also table below).
  • RNA Synthesis
  • Reaction
    Template Nucleotides Volume/μL ivT Yield/μg
    PTH A, G, 5moU, C 15 51.68
  • 100,000 human epidermal keratinocytes (HEKn, Gibco) were plated in EpiLife+Supplement S7. Cells were transfected according to Example 3 with 1 μg of RNA encoding PTH. 24 hours after transfection, the medium was sampled and secreted PTH levels were measured using a human PTH ELISA kit (EIA-PTH-1, Ray Biotech) according the manufacturer's instructions. Secreted PTH levels were determined by measuring 450 nm absorbance using a microplate reader (EMax Plus, Molecular Devices). Secreted PTH levels are shown in FIG. 31.
  • Example 41: Intradermal, Subcutaneous, Rectal and Nasal Administration of NOVECRIT for the Treatment of Anemia
  • A repeat dose toxicity study of Novecrit was conducted for the treatment of anemia. Specifically, 8-10 weeks old male Sprague Dawley rats were administered with Novecrit via intradermal, subcutaneous, rectal, or nasal routes once per day on days 1, 8, and 15. The animals were assigned to groups and treated as indicated in the table below:
  • Dose Dose Number of
    Group Test Concentration Volume Animals
    Group Color Article Dose Route Dose Level (μg) (μg/mL) (μL) Males
    1 White Control Intradermal 0.0 0.0 50 3a + 8b
    4X
    2 Yellow Novecrit Intradermal (0.25 μg/ 5.0 50 3a + 8b
    injection; 1.0 μg
    total)
    3 Green Novecrit Subcutaneous 4.0 20.0 200 3a + 8b
    4 Blue Novecrit Rectal 4.0 20.0 200 3a + 8b
    5 Red Novecrit Nasal 4.0 20.0 200 3a + 8b
    Note:
    Total dose volume (μL/per animal) are constant
    aToxicity animals (also called main study animals), necropsy on Day 44
    bTK animals, euthanized as n = 2/time point on Days 2, 16, 23 and 44
  • More particularly, Group 1 was dosed via intradermal injection. Each dose was administered in four intradermal injections of 50 μL/injection for a total of 200 uL per animal. Injections were carried out on previously marked sites near the midline of the dorsal lumbar area (upper left, upper right, lower left and lower right quadrants). Group 2 was dosed four intradermal injections at 0.25 μg each (1.0 μg total) into previously marked sites near the midline of the dorsal lumbar area (upper left, upper right, lower left and lower right quadrants). Group 3 was dosed by subcutaneous injections into an area of the back located in the lower dorsal thoracic/lumbar region. Group 4 was dosed by rectal administration. The animal was manually restrained and the abdomen of each rat was manually palpated to remove any fecal matter. If deemed necessary, the rectum was lavaged with up to 2 ml of saline for enema followed by manual palpation of the abdomen to remove the fecal matter, if any. Novecrit was drawn into a syringe and an appropriately sized gavage needle with a rounded tip (ball) was attached to the syringe. A lubricant jelly was applied to the insertion device to aid with insertion; it was advanced approximately 1 cm into the lumen of the colon and the Novecrit instilled. The rat was then maintained in a head-down position for approximately 20-30 seconds to limit the expulsion of solution. Group 5 was dosed via nasal route. The animal was anesthetized (per SNBL USA SOP). The animal laid on its back with the head elevated. The dose was dispensed slowly into the nares. Approximately half the dose volume was administered to one nare. The remaining dose volume was administered to another nare. The first dose was administered on Day 1, and the last dose on Day 15.
  • The rats were clinically monitored including their food consumption and body weight. In addition, blood was collected for hematology, coagulation, and serum chemistry analysis as indicated below:
  • Toxicity Group Specimen Collection Frequency
  • Toxicity Group Specimen collection frequency
    Time Point Hematology Coagulation Serum Chemistry
    Day
    44 1X 1X 1X
    (n = 3/Group) (n = 3/Group (n = 3/Group)
    X = Number of times procedure performed
  • Analysis was done on the toxicokinetic group as follows:
  • In-Text Table 3: Toxicokinetic Group Specimen Collection Frequency
  • Time Point Hematologya TK Cytokines
    Acclimation 1X
    (n = 8/Group)
    Day 2 1X 1X
    (n = 2/Group, (n = 2/Group,
    24 hours postdose) 24 hours postdose)
    Day 8 Pre-dose
    (n = 6/Group)
    Day 15 Pre-dose
    (n = 6/Group)
    Day 16 1X 1X
    (n = 2/Group, (n = 2/Group,
    24 hours postdose) 24 hours postdose)
    Day 22 1X
    (n = 4/Group)
    Day 23 1X 1X
    (n = 2/Group) (n = 2/Group)
    Day 29 1X
    (n = 2/Group)
    Day 36 1X
    (n = 2/Group)
    Day 43 1X
    (n = 2/Group)
    Day 44 1X 1X
    (n = 2/Group) (n = 2/Group)
    X = Number of times procedure performed
    TK = toxicokinetics
  • Terminal necropsy for toxicity animals occurred on Day 44. TK animals were euthanized on Days 2, 16, 23, and 44. Pathological analysis was conducted on the animals.
  • As shown in FIG. 32, administration of NOVECRIT stimulated erythropoiesis resulting in elevated hematocrit for at least 14 days in all four study groups compared to control.
  • Example 42: Intradermal Administration of RNA Encoding BMP7 Variants for the Treatment of Diabetic Nephropathy
  • A ZDSD rat model was utilized to study the effects of RNAs encoding BMP7 variants for the prevention and treatment of diabetic nephropathy. Specifically, ZDSD rats were treated with RNAs encoding BMP7 variants administered intradermally. A schematic of the study design is provided in FIG. 33. The animals were assigned to groups and treated as indicated in the table below:
  • Dose Dose Route
    Group No. of Dose Level Conc. Volume of
    Number Treatment Animals (μg) (μg/ml) (ml/kg) Frequency Admin.
    1 Vehicle 24 0 0 0.1 2x/wk I.D.
    2 FTB-F1 (wt 6 1 5 0.1 2x/wk I.D.
    BMP7)
    3 FTB-F2 (BMP7 6 1 5 0.1 2x/wk I.D.
    variant A)
    4 FTB-F3 (BMP7 6 1 5 0.1 2x/wk I.D.
    variant B)
    5 Lisinopril 6 Standard Standard 5 Continuous Diet
    6a Vehicle 20 0 0 0.2/rat 3x/wk I.D.
    7a FTB-F2 10 2 10  0.2/rat 3x/wk I.D.
    (BMP7
    variant A)
    a Group 1 animals at the end of the “Prevention” arm were randomized into Groups 6 and 7 (n = 20 or 10 each, respectively) to define the “Treatment” arm. The remaining 6 animals from Group 1 were euthanized for kidney collection and blood draw at the end of the “Prevention” arm.
  • To study the effect of the RNAs on prevention of diabetic nephropathy, animals were delivered from the barrier (PCO-5638) to the vivarium (PCO-Rm C) and allowed 3 days for acclimation. All animals were placed on 5SCA diabetogenic diet for 3 weeks (weekly body weight measurements were taken). Animals were then returned to regular 5008 diet for the duration of the study (weekly body weight measurements were taken). After 2 weeks on 5008 diet, body weight measurement, blood draw (500 ul) and a 24 hour baseline urine collection (urinary albumin and creatinine measurements) were performed on all animals. 36 rats were randomized to groups 1, 2 and 3 based on body weight, glucose and urinary albumin. Groups 1, 2, 3 and 4 received either vehicle or test article (i.d.) via intradermal administration (2×/week, Mon and Thur). Group 5 was administered Lisinopril admixed in the 5008 diet (food consumption will begin on this group of animals). Blood draws (500 ul, tail vein) were taken from Groups 2, 3 and 4, 24 hours after each dose for the first 2 weeks and then 1×/week thereafter. After 4 weeks of dosing, blood sample via tail vein (500 ul) was taken from all animals (Groups 1-5). At the end of 8 weeks of dosing, a 24 hour urine collection was performed (urinary albumin and creatinine measurements) together with a tail vein (500 ul) blood sample from Group 1 (n=18 going into the Treatment arm). Results for urine volume, creatinine, and albumin is shown in FIG. 35. As shown in FIG. 35, treatment with RNA encoding BMP7 Variant A resulted in reduced levels of urine albumin in rats afflicted with diabetic nephropathy compared to the control, indicating superior kidney function in the treated animals compared to the control animals (placebo group). To terminate the study, all animals were euthanized with CO2 asphyxiation and a blood draw was performed via cardiac puncture to collect serum for BUN, creatinine and glucose, and plasma for compound exposure and biomarker analysis. Both kidneys were dissected and fixed for histology. FIG. 34 shows the serum level of BMP7 protein in Groups 1, 2, 3, and 4. Results indicate that the serum levels of BMP7 protein in these groups were as follows: Group 4, Group 3>Group 2, Group 1.
  • A study was also conducted to analyze the effect of the RNAs encoding BMP7 variants on treating diabetic nephropathy. Specifically, the vehicle animals from Group 1 (Prevention arm, n=18) were randomized based on body weight, glucose and urine albumin (Groups 6, n=20; 7, n=10;). Animals received either Vehicle or BMP7 variant-encoding RNAs (intradermal, 2×/week, Mon and Thur), groups 6 and 7, respectively. After 4 weeks of treatment, animals were euthanized with CO2 asphyxiation and a blood draw will be performed via cardiac puncture to collect serum for BUN, creatinine and glucose, and plasma for compound exposure and biomarker analysis. Both kidneys were dissected and fixed for histology. As shown in FIG. 36, treatment with RNA encoding BMP7 Variant A decreased urine albumin level in rats afflicted with diabetic nephropathy, indicating superior kidney function in the treated animals. This result suggested that BMP7 variant-encoding RNA effectively treated diabetic nephropathy and promoted superior kidney function compared to the control animals (placebo group).
  • An illustrative study protocol is provided below:
  • Week Day Procedure
    15 weeks −7 Animals arrive in Room C from the barrier
    old
    0 BW*, Stat Strip, and Place animals on 5SCA for 3
    wks
    1 7 BW*, Stat Strip
    2 14 BW*, Stat Strip
    3 21 BW*, Stat Strip, put animals on 5008
    4 28 BW*, Stat Strip
    34 BW*, Tail vein blood draw and randomize for
    Prevention arm (Grps 1, 2, 3, 4, 5)
    5 35 Begin 24 hr urine collection
    36 Collect 24 hr urine
    39 BW*
    Intradermal dosing Grps 1, 2, 3 and 4, and put Grp
    5 on admixed 5008 diet
    Begin food consumption on group 5
    40 Collect 24 blood sample for exposure (gps 2, 3, & 4)
    42 Dosing
    43 Collect 24 blood sample for exposure (gps 2, 3, & 4)
     6 (1 P) 46 BW*, Dosing
    Food consumption group 5
    47 Collect 24 blood sample for exposure (gps 2, 3, & 4)
    49 Dosing
    50 Collect 24 blood sample for exposure (gps 2, 3, & 4)
     7 (2 P) 53 BW*, Dosing
    Food consumption group 5
    54 Collect 24 blood sample for exposure (gps 2, 3, & 4)
    56 Dosing
     8 (3 P) 60 BW*, Dosing
    Food consumption group 5
    61 Collect 24 blood sample for exposure (gps 2, 3, & 4)
    63 Dosing
     9 (4 P) 67 BW*, Dosing
    Food consumption group 5
    Blood draw (tail vein)
    68 Blood draw (all groups)
    Collect 24 blood sample for exposure (gps 2, 3, & 4)
    70 Dosing
    10 (5 P) 74 BW*, Dosing
    Food consumption group 5
    75 Collect 24 blood sample for exposure (gps 2, 3, & 4)
    77 Dosing
    11 (6 P) 81 BW*, Dosing
    Food consumption group 5
    Collect 24 blood sample for exposure (gps 2, 3, & 4)
    84 Dosing
    12 (7 P) 88 BW*, Dosing
    Food consumption group 5
    89 Collect 24 blood sample for exposure (gps 2, 3, & 4)
    91 Dosing
    13 (8 P) 95 BW*, dosing
    Food consumption group 5
    Blood draw (tail vein) (all groups)
    96 Begin 24 hr urine collection
    97 Collect 24 hr urine
    Terminate Groups 1 (n = 6), 2, 3, 4 and 5 cardiac
    puncture blood draw, and collect kidneys, weigh and
    fix.
    98 BW* and randomize (BW*, glucose, albumin) Group
    1 (n = 18) rats into Groups 6, 7 and 8
    Dose
    Food Consumption group 8
    103 Dose
    14 (1 T) 106 BW*, Dosing
    Food Consumption group 8
    110 Dosing
    15 (2 T) 113 BW*, Dosing
    Food Consumption group 8
    117 Dosing
    16 (3 T) 120 BW*, Dosing
    Food Consumption group 8
    124 Dosing
    17 (4 T) 127 BW*, Terminate, cardiac puncture blood draw, and
    collect kidneys, weigh and fix.
    *BW: body weight measurement
  • In an illustrative study, the protocol was amended from above towards the end of the study as follows:
  • Treatment BW
    Arm Day Begin 24 hr urine collection
    81 Collect 24 hr urine
    BW, Blood draw
    Randomize (BW, glucose, albumin) rats into Groups 6 and 7
    1 T BW, Dose
    12 hr PK sample
    Dose
    Dose and Begin 24 hr Urine collection
    89 Collect 24 hr Urine and Dose
    2T BW, Dose
    12 hr PK sample
    Dose
    Begin
    24 hr Urine collection
    96 Collect 24 hr Urine and Dose
    3T BW, Dose
    12 hr PK sample
    Dose
    Begin
    24 hr Urine collection
    Collect 24 hr Urine and Dose (last dose)
    4T BW
    No activity
    Begin 24 hr urine collection
    Collect 24 hr urine
    BW, Terminate, cardiac puncture blood draw, and
    collect kidneys, weigh and fix.
  • During both studies, pathological analysis is conducted as follows:
  • Clinical
    Specimen to Method of Anti- Specimen to Chemistry Method of
    be Collected Collection Coagulant be Analyzed Analyte Analysis
    Urine 24hr NA Urine Creatinine/ AU480/
    Metabolism Albumin ICL Elisa
    Cages
    Whole Blood Tail Vein None Serum BUN AU480/
    Creatinine Sponsor
    Glucose/
    Whole Blood Cardiac None Serum BUN AU480/
    Puncture Creatinine Sponsor
    ( Grps 1,2, 3, Glucose/
    4 and 5; and Cmpd Exp &
    6, 7, 8) Biomarker
  • Altogether, these results suggested that RNAs encoding BMP-7 variants effectively prevented the development of diabetic nephropathy in rats. In addition, these RNAs also effectively treated and restored kidney functions in rats already afflicted with diabetic nephropathy. RNA encoding BMP7 Variant A was particularly effective at preventing and treating diabetic nephropathy in rats.
  • Example 43: Transfection of Human Keratinocytes with RNA Encoding BDNF, BMP-2, BMP-6, IL-2, IL-6, IL-15, IL-22, LIF or FGF-21
  • 100,000 human epidermal keratinocytes (HEK, Gibco) were plated in EpiLife+Supplement S7. Cells were transfected according to Example 3 with 2 μg of RNA encoding BDNF, BMP-2, BMP-6, IL-2, IL-6, IL-15, IL-22, LIF or FGF-21. 24 hours after transfection, the medium was sampled and secreted protein levels were measured using a human ELISA kit (see Table below) according the manufacturer's instructions. Secreted protein levels were determined by measuring 450 nm absorbance using a microplate reader (EMax Plus, Molecular Devices). Secreted protein levels are shown in FIG. 34, panels A-I.
  • Protein Part Number Vendor
    BDNF DBNT00 R&D Systems
    BMP-2 DBP200 R&D Systems
    BMP-6 ab99984 Abcam
    IL-2 D2050 R&D Systems
    IL-6 D6050 R&D Systems
    IL-15 D1500 R&D Systems
    IL-22 D2200 R&D Systems
    LIF DLF00 R&D Systems
    FGF-21 DF2100 R&D Systems
  • Example 44: Transfection of Human Keratinocytes with RNA Encoding IL-15 and/or IL-15RA
  • 100,000 human epidermal keratinocytes (HEK, Gibco) were plated in EpiLife+Supplement S7. Cells were transfected according to Example 3 with 2 μg of RNA encoding IL-15 or 1 μg each of RNA encoding IL-15 and RNA encoding IL-15RA. 24 hours after transfection, the medium was sampled and secreted IL-15 levels were measured using a human IL-15 ELISA kit (D1500, R&D Systems) according the manufacturer's instructions. Secreted IL-15 levels were determined by measuring 450 nm absorbance using a microplate reader (EMax Plus, Molecular Devices). Secreted IL-15 levels are shown in FIG. 35. As demonstrated in FIG. 35, co-transfection of IL-15 and IL-15RA significantly increased secreted IL-15 levels compared to transfection with IL-15 alone.
  • Example 45: Pharmacokinetic Study Via Intradermal Injection in Rats
  • Studies were conducted to evaluate the responses of Sprague Dawley rats to intradermal administration of various RNAs. Specifically, 8-10 weeks old, female Sprague Dawley rats weighing about 200 g to about 350 g were used for this study. A total of 33 rats were tested, and the animals were assigned to study groups and treated as indicated in the Table below:
  • Dose
    Dose Volume Number of
    Test Dose Dose Level Concentration (μL/per Animals
    Group Group Color Article Route (μg) (μg/mL) injection)a Females
    1 White Control ID 4.0 20.0 200 3b
    (NOVEPOEITIN) (4 × 50)
    2 Yellow TA1 ID 4.0 20.0 200 3b
    (IL2) (4 × 50)
    3 Green TA2 ID 4.0 20.0 200 3b
    (IL6) (4 × 50)
    4 Blue TA3 ID 4.0 10.0 200 3b
    (IL15) (4 × 50)
    5 Red TA4 ID 4.0 20.0 200 3b
    (IL15 + (10.0 each) (4 × 50)
    IL15RA)
    6 Dark Grey TA5 ID 4.0 20.0 200 3b
    (IL22) (4 × 50)
    7 Purple TA6 ID 4.0 20.0 200 3b
    (BMP2) (4 × 50)
    8 Black TA7 ID 4.0 20.0 200 3b
    (BDNF) (4 × 50)
    9 White/Yellow TA8 ID 4.0 20.0 200 3b
    (LIF) (4 × 50)
    10 Green/Blue TA9 ID 4.0 20.0 200 3b
    (PTH) (4 × 50)
    11 Red/Dark Grey TA10 ID 4.0 20.0 200 3b
    (FGF21) (4 × 50)
    aTotal dose volume (μL/per animal) was constant. Each animal received four intradermal injections of 50 μL/per injection for a total of 200 μL per animal.
    bAnimals, euthanized on Day 3 without examination or necropsy
    Intradermal = ID
  • For the study, the animals were treated with either 4 ug of RNA. All groups were dosed via intradermal injection. Each dose was administered in four intradermal injections of 50 μL/injection for a total of 200 uL per animal. Injections occurred into previously marked sites near the midline of the dorsal lumbar area (upper left, upper right, lower left and lower right quadrants). Dose time (after the last injection) was recorded. Additional markings were made as needed to allow for identification of the dose site. Animals were administered with the RNAs on day 1 and euthanized on day 3. Clinical observations were made on the rats twice daily. Food consumption and body weight were also monitored.
  • During the study, approximately 1 ml of blood samples was collected from the jugular vein for pharmacokinetic analysis as follows:
  • Time Point PKa
    Acclimation
    Day 1 12 hours postdose
    Day 2 24 hours postdose
    Day 3 48 hours postdose
    aTime points for blood collection (n = 3 animals/Group/Time point)
    PK = Pharmacokinetics
  • Results indicate that following administration of RNAs encoding FGF21, IL15, IL15 and IL15R, IL6, IL22, and NOVEPOEITIN, these proteins were readily detected in the blood with protein levels peaking at approximately 12 hours post injection (FIG. 39). Of note, the proteins tested in this study can be taken up by cells and tissues and/or can exert an effect near the site of expression without appreciable accumulation in systemic circulation.
  • EQUIVALENTS
  • Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
  • INCORPORATION BY REFERENCE
  • All patents and publications referenced herein are hereby incorporated by reference in their entireties.

Claims (20)

1.-64. (canceled)
65. A method for treating cancer comprising:
(a) providing an isolated cell;
(b) transfecting the cell with a synthetic RNA encoding a catalytic enzyme capable of converting a prodrug into an active drug, to result in the cell expressing the catalytic enzyme;
(c) administering the cell to a cancer patient; and
(d) administering the prodrug to the cancer patient, to result in conversion of the prodrug into an active drug.
66. The method of claim 65, wherein the prodrug is selected from the group consisting of 5-Ethynyl-2(1H)-pyrimidinone, IPdR, 5-FP, d-alanine, SeCys conjugates, Menadione, Mitomycin C, Tirapazamine, EO9, Streptonigrin, CB 1954, Diaziquone, Ipomeanol, Ftorafur (tegafur), 5′-DFUR, Dacarbazine, Trofosfamide, Ifosfamide, Cyclophosphamide, AQ4N, 2,4-Dihydroxyphenylalanine, 4-S-CAP, GHB, Substituted phenols, Phenyl mustards, Urea mustards, TER286, S-CPHC-ethylsulfoxide, PTA, CPT-11, Paclitaxel-2-ethylcarbonate, Capecitabine, Tertiary amidomethyl esters, Amifostine, 3-AP phosphate, BHAMG, Epirubicin-glucuronide, HMR 1826, DNR-GA3, DOX-GA3, Paclitaxel glucuronide, 5-FU glucuronide, 5-FC, PC, GC, SeCys conjugate, 4-Ipomeanol, Fludarabine, MeP-dR, Ganciclovir, Etoposide phosphate, Mitomycin C phosphate, POMP, N-(4-phosphonooxy)-phenylacetyl)doxorubicin, Glucuronidated Nornitrogen mustard, Glucuronidated 9-amino-camptothecin, Glucuronide mustard, Methotrexate-amino acids, CMDA, DPO, N-(phenyacetyl) doxorubicin, MelPO, NHPAP, N-(phenylacetyl) doxorubicin, N-(phenylacetyl) melphalan, LY 266070, C-DOX, PRODOX, CM, CCM, Cephalosporin-DACCP, PROTAX, Cephalosporin mitomycin C, C-Mel, 5-Fluorocytosine, Selenomethionine, and Trifluoromethionine.
67. The method of claim 66, wherein the catalytic enzyme is selected from the group consisting of an aldehyde oxidase, an amino acid oxidase, a cytochrome P450 reductase, a DT-diaphorase, a cytochrome P450, a tyrosinase, a glutathione an S-transferase, a carboxylesterase, an alkaline phosphatase, a β-glucuronidase, a cysteine conjugate β-lyase, a nitroreductase, a purine-nucleoside phosphorylase, a thymidine phosphorylase, a thymidine kinase, a carboxypeptidase, a penicillin amidase, a β-Lactamase, a cytosine deaminase, and a methionine γ-lyase.
68. The method of claim 65, wherein the catalytic enzyme is capable of catalyzing conversion of the prodrug selected from the group consisting of 5-FP, tegafur, 5′-DFUR, 5-FU glucuronide, and 5-FC, into the active drug 5-FU, and wherein the catalytic enzyme is selected from the group consisting of an aldehyde oxidase, a cytochrome P450, a thymidine phosphorylase, a β-glucuronidase, and a cytosine deaminase.
69. The method of claim 68, wherein the catalytic enzyme comprises the cytosine deaminase, and the cytosine deaminase is capable of catalyzing conversion of 5-FC into 5-FU.
70. The method of claim 68, wherein the catalytic enzyme comprises the aldehyde oxidase that is capable of catalyzing conversion of 5-FC into 5-FU, or the catalytic enzyme comprises the cytochrome P450 that is capable of catalyzing conversion of tegafur into 5-FU, or the catalytic enzyme comprises a carboxylesterase that is capable of catalyzing conversion of a capecitabine into 5-FU, or the catalytic enzyme comprises the β-glucuronidase that is capable of catalyzing conversion of 5-FU glucoronide into 5-FU.
71. The method of claim 67, wherein the catalytic enzyme is capable of catalyzing conversion of the prodrug selected from the group consisting of N-(4-phosphono-oxy)-phenylacetyl) doxorubicin, HMR 1826, DOX-GA3, DPO, N-(phenyacetyl) doxorubicin, C-DOX, and PRODOX into the active drug 5-FU, and wherein the catalytic enzyme is selected from the group consisting of the alkaline phosphatase, the β-glucuronidase, the penicillin amidase, and the β-Lactamase.
72. The method of claim 65, wherein the synthetic RNA comprises a combination of non-canonical nucleotides that avoid substantial cellular toxicity, optionally selected from 5-hydroxycytidine, 5-methylcytidine, 5-hydroxymethylcytidine, 5-carboxycytidine, 5-formylcytidine, 5-methoxycytidine, pseudouridine, 5-hydroxyuridine, 5-methyluridine, 5-hydroxymethyluridine, 5-carboxyuridine, 5-formyluridine, 5-methoxyuridine, 5-hydroxypseudouridine, 5-methylpseudouridine, 5-hydroxymethylpseudouridine, 5-carboxypseudouridine, 5-formylpseudouridine, and 5-methoxypseudouridine, optionally at an amount of at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% of the non-canonical nucleotides.
73. The method of claim 65, wherein the synthetic RNA comprises a 5′ cap structure.
74. The method of claim 65, wherein the synthetic RNA comprises a 5′-UTR comprising a Kozak consensus sequence, the 5′-UTR optionally comprises a sequence that increases RNA stability in vivo, and the 5′-UTR optionally comprises an alpha-globin or beta-globin 5′-UTR.
75. The method of claim 65, wherein the synthetic RNA comprises a 3′-UTR comprising a sequence that increases RNA stability in vivo, and the 3′-UTR optionally comprises an alpha-globin or beta-globin 3′-UTR.
76. The method of claim 65, wherein the synthetic RNA comprises a 3′ poly(A) tail, optionally from about 20 nucleotides to about 250 nucleotides in length.
77. The method of claim 65, wherein the prodrug is administered systemically.
78. The method of claim 65, wherein the cancer is selected from the basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulval cancer; lymphoma including Hodgkin's and non-Hodgkin's lymphoma, as well as B-cell lymphoma (including low grade/follicular non-Hodgkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; and Waldenstrom's Macroglobulinemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); Hairy cell leukemia; chronic myeloblastic leukemia; as well as other carcinomas and sarcomas; and post-transplant lymphoproliferative disorder (PTLD), as well as abnormal vascular proliferation associated with phakomatoses, edema (such as that associated with brain tumors), and Meigs' syndrome.
79.-84. (canceled)
85. The method of claim 65, wherein the cell is a skin cell.
86. The method of claim 85, wherein the skin cell is selected from the group consisting of a fibroblast, a keratinocyte, a melanocyte, an adipocyte, a mesenchymal stem cell, an adipose stem cell, and a blood cell.
87. The method of claim 65, further comprising transfecting the cell with a synthetic RNA encoding a targeting molecule capable of directing the cell to a tumor, to result in the cell expressing the targeting molecule.
88. The method of claim 77, wherein the prodrug is administered by injection.
US16/789,831 2015-02-13 2020-02-13 Nucleic acid products and methods of administration thereof Pending US20200261599A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/789,831 US20200261599A1 (en) 2015-02-13 2020-02-13 Nucleic acid products and methods of administration thereof

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201562116232P 2015-02-13 2015-02-13
US201562195462P 2015-07-22 2015-07-22
US201562242383P 2015-10-16 2015-10-16
US201562245726P 2015-10-23 2015-10-23
US201662289617P 2016-02-01 2016-02-01
PCT/US2016/018065 WO2016131052A1 (en) 2015-02-13 2016-02-16 Nucleic acid products and methods of administration thereof
US201715550280A 2017-08-10 2017-08-10
US16/789,831 US20200261599A1 (en) 2015-02-13 2020-02-13 Nucleic acid products and methods of administration thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2016/018065 Continuation WO2016131052A1 (en) 2015-02-13 2016-02-16 Nucleic acid products and methods of administration thereof
US15/550,280 Continuation US11241505B2 (en) 2015-02-13 2016-02-16 Nucleic acid products and methods of administration thereof

Publications (1)

Publication Number Publication Date
US20200261599A1 true US20200261599A1 (en) 2020-08-20

Family

ID=56615771

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/550,280 Active US11241505B2 (en) 2015-02-13 2016-02-16 Nucleic acid products and methods of administration thereof
US16/789,831 Pending US20200261599A1 (en) 2015-02-13 2020-02-13 Nucleic acid products and methods of administration thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/550,280 Active US11241505B2 (en) 2015-02-13 2016-02-16 Nucleic acid products and methods of administration thereof

Country Status (7)

Country Link
US (2) US11241505B2 (en)
EP (2) EP3543339A1 (en)
JP (2) JP7199809B2 (en)
CN (1) CN109477102A (en)
AU (2) AU2016218977C1 (en)
CA (1) CA2976376A1 (en)
WO (1) WO2016131052A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11904023B2 (en) 2016-08-17 2024-02-20 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6728156B2 (en) 2014-11-02 2020-07-22 アークトゥラス・セラピューティクス・インコーポレイテッドArcturus Therapeutics,Inc. Messenger UNA molecule and its use
WO2018104540A1 (en) * 2016-12-08 2018-06-14 Curevac Ag Rnas for wound healing
MX2019009117A (en) 2017-02-17 2019-09-13 Bristol Myers Squibb Co Antibodies to alpha-synuclein and uses thereof.
EP3585897A1 (en) * 2017-02-22 2020-01-01 CRISPR Therapeutics AG Materials and methods for treatment of dystrophic epidermolysis bullosa (deb) and other collagen type vii alpha 1 chain (col7a1) gene related conditions or disorders
CN110636854A (en) * 2017-03-31 2019-12-31 阿卡尼斯生物技术F&E有限责任两合公司 Prevention and treatment of non-melanoma skin cancer (NMSC)
EP3437650A1 (en) 2017-07-31 2019-02-06 Accanis Biotech F&E GmbH & Co KG Treatment of local skin hypotrophy conditions
WO2019158745A1 (en) * 2018-02-16 2019-08-22 Bontana Therapies Gmbh Nucleic acid-based botulinum neurotoxin for therapeutic use
WO2020146700A1 (en) * 2019-01-10 2020-07-16 Massachusetts Institute Of Technology Lipid nanoparticles
WO2020223674A2 (en) * 2019-05-01 2020-11-05 Board Of Regents Of The University Of Texas System Methods and compositions for regulating adipogenesis
CN114450265A (en) * 2019-07-03 2022-05-06 菲克特生物科学股份有限公司 Cationic lipids and uses thereof
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
WO2021030268A2 (en) 2019-08-09 2021-02-18 Nutcracker Therapeutics, Inc. Microfluidic apparatus and methods of use thereof
WO2021231549A2 (en) * 2020-05-12 2021-11-18 Factor Bioscience Inc. Engineered gene-editing proteins
CN114717229A (en) * 2021-01-05 2022-07-08 麦塞拿治疗(香港)有限公司 Cell-free and vector-free in vitro RNA transcription methods and nucleic acid molecules for therapeutic mRNA
CN114350801B (en) * 2022-01-05 2023-10-20 华北理工大学 mRNA detection primer set, probe, kit and application for early screening of pneumoconiosis patient
CN116675780A (en) * 2023-06-02 2023-09-01 东北农业大学 Preparation of canine fibroblast growth factor21 fusion protein and application thereof in treating atopic dermatitis
CN117783501A (en) * 2024-02-26 2024-03-29 四川大学华西第二医院 Application of sodium octoate in attention deficit and hyperactivity disorder and product

Family Cites Families (165)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3034507A (en) 1960-05-10 1962-05-15 American Cyanamid Co Intracutaneous injection device
US3539465A (en) 1968-10-08 1970-11-10 Ncr Co Encapsulation of hydrophilic liquid-in-oil emulsions
US3675766A (en) 1970-02-04 1972-07-11 Sol Roy Rosenthal Multiple puncture injector device
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US6800603B2 (en) 1991-03-11 2004-10-05 Curis, Inc. Morphogen-induced neural cell adhesion
US6949505B1 (en) 1991-03-11 2005-09-27 Curis, Inc. Morphogen-induced dendritic growth
US5849686A (en) 1991-03-11 1998-12-15 Creative Biomolecules, Inc. Morphogen-induced liver regeneration
US5674844A (en) 1991-03-11 1997-10-07 Creative Biomolecules, Inc. Treatment to prevent loss of and/or increase bone mass in metabolic bone diseases
US5656593A (en) 1991-03-11 1997-08-12 Creative Biomolecules, Inc. Morphogen induced periodontal tissue regeneration
US6506729B1 (en) 1991-03-11 2003-01-14 Curis, Inc. Methods and compositions for the treatment and prevention of Parkinson's disease
US5993434A (en) 1993-04-01 1999-11-30 Genetronics, Inc. Method of treatment using electroporation mediated delivery of drugs and genes
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5711964A (en) 1995-06-07 1998-01-27 United States Of America Method for the intracellular delivery of biomolecules using liposomes containing cationic lipids and vitamin D
WO1997034618A1 (en) 1996-03-22 1997-09-25 The General Hospital Corporation Administration of polypeptide growth factors following central nervous system ischemia or trauma
US6498142B1 (en) 1996-05-06 2002-12-24 Curis, Inc. Morphogen treatment for chronic renal failure
US6602693B1 (en) 1996-07-03 2003-08-05 Clear Solutions Biotech, Inc. Gene encoding hyaluronan synthase
US6770291B2 (en) 1996-08-30 2004-08-03 The United States Of America As Represented By The Department Of Health And Human Services Liposome complexes for increased systemic delivery
JP4203835B2 (en) 1996-08-19 2009-01-07 アメリカ合衆国 Novel liposomal complexes that increase systemic delivery
TW520297B (en) 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
US6653104B2 (en) 1996-10-17 2003-11-25 Immunomedics, Inc. Immunotoxins, comprising an internalizing antibody, directed against malignant and normal cells
JP2001508302A (en) 1997-01-10 2001-06-26 ライフ テクノロジーズ,インコーポレイテッド Embryonic stem cell serum replacement
US6316260B1 (en) 1997-08-22 2001-11-13 Bernina Biosystems Gmbh Tetraether lipid derivatives and liposomes and lipid agglomerates containing tetraether lipid derivatives, and use thereof
WO1999014346A2 (en) 1997-09-19 1999-03-25 Sequitur, Inc. SENSE mRNA THERAPY
DK1129064T3 (en) 1998-11-12 2008-04-28 Invitrogen Corp transfection
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
EP1187653B1 (en) 1999-06-04 2010-03-31 Georgia Tech Research Corporation Devices for enhanced microneedle penetration of biological barriers
US20110171185A1 (en) 1999-06-30 2011-07-14 Klimanskaya Irina V Genetically intact induced pluripotent cells or transdifferentiated cells and methods for the production thereof
US20020054895A1 (en) 1999-07-23 2002-05-09 Alwyn Company, Inc. Allantoin-containing skin cream
US6595947B1 (en) 2000-05-22 2003-07-22 Becton, Dickinson And Company Topical delivery of vaccines
AU9475001A (en) 2000-09-26 2002-04-08 Hybridon Inc Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
US7189759B2 (en) 2001-05-23 2007-03-13 Medicis Pharmaceutical Corporation Compositions for the treatment of pigmentation disorders and methods for their manufacture
AU2002313817A1 (en) 2001-08-27 2003-03-10 Advanced Cell Technology, Inc. Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
WO2003027277A1 (en) 2001-09-21 2003-04-03 Japan Science And Technology Corporation Method of screening reporgramming factor, reprogramming factor screened by the method, method of using the reprogramming factor, method of differentiating undifferentiated fused cells and method of constructing cell, tissues and organs
US20030138951A1 (en) 2001-10-18 2003-07-24 Li Yin Conversion of liver stem and progenitor cells to pancreatic functional cells
US7276489B2 (en) 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
GB0202149D0 (en) 2002-01-30 2002-03-20 Univ Edinburgh Pluripotency determining factors and uses thereof
US20030186249A1 (en) 2002-04-01 2003-10-02 Zairen Sun Human TARPP genes and polypeptides
JP3785508B2 (en) 2002-04-15 2006-06-14 学校法人慶應義塾 Experimental model mouse that can analyze immune response in gene therapy
DE10234192B4 (en) * 2002-07-26 2009-11-26 Epoplus Gmbh Co.Kg Use of erythropoietin
US7737182B2 (en) 2002-08-09 2010-06-15 Taisho Pharmaceutical Co., Ltd. Pharmaceuticals for xerosis
EP2241572A3 (en) 2003-06-03 2011-04-06 Eli Lilly And Company Modulation of survivin expression
WO2005051351A2 (en) 2003-11-21 2005-06-09 Alza Corporation Gene delivery mediated by liposome-dna complex with cleavable peg surface modification
US7682828B2 (en) 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
NZ553241A (en) 2004-09-08 2009-11-27 Wisconsin Alumni Res Found Medium and culture of pluripotent stem cells
US7442548B2 (en) 2004-09-08 2008-10-28 Wisconsin Alumni Research Foundation Culturing human embryonic stem cells in medium containing pipecholic acid and gamma amino butyric acid
US20090202638A2 (en) 2004-10-27 2009-08-13 New York Society For The Ruptured And Crippled Maintaining The Hospital For Special Surgery Bmp gene and fusion protein
AU2006208199B2 (en) 2005-01-25 2012-06-07 Cell Therapeutics, Inc. Conjugates of biologically active proteins having a modified in vivo half-life
CN101203222A (en) * 2005-03-30 2008-06-18 艾升制药公司 Dermatological compositions and salts for the treatment of dermatological diseases
ES2397851T3 (en) 2005-07-13 2013-03-11 Novo Nordisk Health Care Ag Host cell protein inactivation cells for the production of therapeutic proteins
CA2615532C (en) 2005-07-26 2016-06-28 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US8323666B2 (en) 2005-08-01 2012-12-04 Allergan, Inc. Botulinum toxin compositions
EP4174179A3 (en) 2005-08-23 2023-09-27 The Trustees of the University of Pennsylvania Rna containing modified nucleosides and methods of use thereof
US9012219B2 (en) 2005-08-23 2015-04-21 The Trustees Of The University Of Pennsylvania RNA preparations comprising purified modified RNA for reprogramming cells
EP1962719A4 (en) 2005-08-29 2011-05-04 Technion Res And Dev Of Foundation Ltd Media for culturing stem cells
TW200732347A (en) 2005-10-06 2007-09-01 Trophogen Inc VEGF analogs and methods of use
ZA200804673B (en) 2005-12-13 2009-11-25 Univ Kyoto Nuclear reprogramming factor
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US20100168000A1 (en) 2005-12-22 2010-07-01 Peter Kiessling Octanoate-Reduced Human Albumin
US7658728B2 (en) 2006-01-10 2010-02-09 Yuzhakov Vadim V Microneedle array, patch, and applicator for transdermal drug delivery
DE102006051516A1 (en) 2006-10-31 2008-05-08 Curevac Gmbh (Base) modified RNA to increase the expression of a protein
GB0623635D0 (en) 2006-11-27 2007-01-03 Stem Cell Sciences Uk Ltd Pluripotent cell growth media
US7785301B2 (en) 2006-11-28 2010-08-31 Vadim V Yuzhakov Tissue conforming microneedle array and patch for transdermal drug delivery or biological fluid collection
AU2007333225B2 (en) 2006-12-08 2014-06-12 Massachusetts Institute Of Technology Delivery of nanoparticles and/or agents to cells
CN101200758A (en) 2006-12-15 2008-06-18 华中科技大学 Method for detecting COL7A1 gene mutation and uses thereof
US10829733B2 (en) 2007-01-04 2020-11-10 Biolamina Ab Composition and method for enabling proliferation of pluripotent human stem cells
JP2010515464A (en) 2007-01-11 2010-05-13 イエール・ユニバーシテイ Compositions and methods for targeted inactivation of HIV cell surface receptors
WO2008097926A2 (en) 2007-02-02 2008-08-14 Yale University Transient transfection with rna
US9249423B2 (en) 2007-02-02 2016-02-02 Yale University Method of de-differentiating and re-differentiating somatic cells using RNA
EP2137296A2 (en) 2007-03-23 2009-12-30 Wisconsin Alumni Research Foundation Somatic cell reprogramming
EP3878949A1 (en) 2007-04-07 2021-09-15 Whitehead Institute for Biomedical Research Reprogramming of somatic cells
JP2008307007A (en) 2007-06-15 2008-12-25 Bayer Schering Pharma Ag Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
EP2206714B1 (en) * 2007-09-28 2015-01-21 Kyowa Hakko Kirin Co., Ltd. Agent for prevention and/or treatment of skin diseases
JP5558097B2 (en) 2007-12-10 2014-07-23 国立大学法人京都大学 Efficient nuclear initialization method
CN101903299B (en) 2007-12-14 2013-03-20 帝斯曼知识产权资产管理有限公司 Sol-gel process with a protected catalyst
EP2072618A1 (en) 2007-12-14 2009-06-24 Johannes Gutenberg-Universität Mainz Use of RNA for reprogramming somatic cells
JP5529754B2 (en) 2007-12-21 2014-06-25 ストライカー コーポレイション BMP variant with reduced sensitivity to Noggin
MX2010010165A (en) 2008-03-17 2010-11-25 Scripps Research Inst Combined chemical and genetic approaches for generation of induced pluripotent stem cells.
US20110045001A1 (en) 2008-03-28 2011-02-24 Biontex Laboratories Gmbh Transfection results of non-viral gene delivery systems by influencing of the innate immune system
WO2009127230A1 (en) 2008-04-16 2009-10-22 Curevac Gmbh MODIFIED (m)RNA FOR SUPPRESSING OR AVOIDING AN IMMUNOSTIMULATORY RESPONSE AND IMMUNOSUPPRESSIVE COMPOSITION
JP2011160661A (en) 2008-06-02 2011-08-25 Kyowa Hakko Kirin Co Ltd Reprogramming method of blood cell
KR101871192B1 (en) 2008-06-04 2018-06-27 후지필름 셀룰러 다이내믹스, 인코포레이티드 Methods for the production of iPS cells using non-viral approach
GB2460552B (en) 2008-06-05 2011-09-07 Iti Scotland Ltd Stem cell culture media and methods
US8669048B2 (en) 2008-06-24 2014-03-11 Parkinson's Institute Pluripotent cell lines and methods of use thereof
US20100184033A1 (en) 2008-07-16 2010-07-22 West Michael D Methods to accelerate the isolation of novel cell strains from pluripotent stem cells and cells obtained thereby
EP2151248A1 (en) 2008-07-30 2010-02-10 Johann Bauer Improved pre-mRNA trans-splicing molecule (RTM) molecules and their uses
US20110263015A1 (en) 2008-08-20 2011-10-27 Virxsys Corporation Compositions and methods for generation of pluripotent stem cells
US20100076057A1 (en) 2008-09-23 2010-03-25 Northwestern University TARGET DNA INTERFERENCE WITH crRNA
CN105802917A (en) 2008-10-24 2016-07-27 威斯康星校友研究基金会 Pluripotent stem cells obtained by non-viral reprogramming
CN105999538A (en) 2008-11-18 2016-10-12 3M创新有限公司 Hollow microneedle array and method
EP2192174B1 (en) 2008-11-21 2015-11-11 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Reprogramming cells toward a pluripotent state
US20110239315A1 (en) 2009-01-12 2011-09-29 Ulla Bonas Modular dna-binding domains and methods of use
EP2206723A1 (en) 2009-01-12 2010-07-14 Bonas, Ulla Modular DNA-binding domains
EP2412800A1 (en) 2010-07-29 2012-02-01 Koninklijke Nederlandse Akademie van Wetenschappen Liver organoid, uses thereof and culture method for obtaining them
US20100209404A1 (en) 2009-02-10 2010-08-19 University Of Dayton Enhanced method for producing stem-like cells from somatic cells
US10894944B2 (en) 2009-04-10 2021-01-19 Monash University Cell culture media
EP3904505A1 (en) 2009-04-22 2021-11-03 Viacyte, Inc. Cell compositions derived from dedifferentiated reprogrammed cells
EP2421563B1 (en) 2009-04-22 2017-04-12 Massachusetts Institute of Technology Innate immune suppression enables repeated delivery of long rna molecules
US10837020B2 (en) 2009-04-22 2020-11-17 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long RNA molecules
EP2251437B1 (en) 2009-05-13 2013-12-04 Hannelore Breitenbach-Koller Method for identifying compounds that control the translational activity of ribosomal proteins in differential mRNA expression
US8496941B2 (en) 2009-06-03 2013-07-30 National Institute Of Advanced Industrial Science And Technology Vectors for generating pluripotent stem cells and methods of producing pluripotent stem cells using the same
EP2443237B1 (en) 2009-06-16 2017-02-22 CuRNA, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
DK2459231T3 (en) 2009-07-31 2016-09-05 Ethris Gmbh RNA with a combination of unmodified and modified nucleotides for protein expression
CA2777710C (en) 2009-11-04 2021-02-23 Cellular Dynamics International, Inc. Episomal reprogramming with chemicals
AU2010319555A1 (en) 2009-11-11 2012-06-14 Sanford-Burnham Medical Research Institute Method for generation and regulation of iPS cells and compositions thereof
US20130189741A1 (en) 2009-12-07 2013-07-25 Cellscript, Inc. Compositions and methods for reprogramming mammalian cells
KR102505097B1 (en) 2009-12-07 2023-03-02 더 트러스티스 오브 더 유니버시티 오브 펜실베니아 Rna preparations comprising purified modified rna for reprogramming cells
KR102110725B1 (en) 2009-12-10 2020-05-13 리전츠 오브 더 유니버스티 오브 미네소타 Tal effector-mediated dna modification
US8557972B2 (en) 2009-12-21 2013-10-15 University Of Washington Through Its Center For Commercialization Compositions and methods for transfection of RNA and controlled stabilization of transfected RNA
US8962331B2 (en) 2010-02-01 2015-02-24 The Board Of Trustees Of The Leland Stanford Junior University Method of making induced pluripotent stem cell from adipose stem cells using minicircle DNA vectors
US20140194482A1 (en) 2010-02-02 2014-07-10 Scioderm, Inc. Compositions and methods of treatment of inflammatory skin conditions using allantoin
JPWO2011102532A1 (en) 2010-02-16 2013-06-17 国立大学法人九州大学 Induced hepatocytes
WO2011110886A1 (en) 2010-03-09 2011-09-15 Biolamina Ab Composition and method for enabling proliferation of pluripotent human stem cells
US8802438B2 (en) 2010-04-16 2014-08-12 Children's Medical Center Corporation Compositions, kits, and methods for making induced pluripotent stem cells using synthetic modified RNAs
CN102234627B (en) 2010-04-30 2015-06-03 中国科学院广州生物医药与健康研究院 Culture medium additive and application thereof
WO2011140397A2 (en) 2010-05-05 2011-11-10 The Regents Of The University Of California Office Of The President Stem cell defined media for xeno-free and feeder free conditions and uses thereof
US8048675B1 (en) 2010-05-12 2011-11-01 Ipierian, Inc. Integration-free human induced pluripotent stem cells from blood
EP2569424A1 (en) 2010-05-12 2013-03-20 Cellectis Meganuclease variants cleaving a dna target sequence from the dystrophin gene and uses thereof
AU2011256838B2 (en) 2010-05-17 2014-10-09 Sangamo Therapeutics, Inc. Novel DNA-binding proteins and uses thereof
EP2392208B1 (en) 2010-06-07 2016-05-04 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Fusion proteins comprising a DNA-binding domain of a Tal effector protein and a non-specific cleavage domain of a restriction nuclease and their use
WO2011159369A1 (en) 2010-06-14 2011-12-22 Iowa State University Research Foundation, Inc. Nuclease activity of tal effector and foki fusion protein
KR101829488B1 (en) 2010-08-05 2018-02-14 위스콘신 얼럼나이 리서어치 화운데이션 Simplified basic media for human pluripotent cell culture
EP3578205A1 (en) 2010-08-06 2019-12-11 ModernaTX, Inc. A pharmaceutical formulation comprising engineered nucleic acids and medical use thereof
WO2012021632A2 (en) 2010-08-10 2012-02-16 The Johns Hopkins University Generation and use of pluripotent stem cells
EP2616540A4 (en) 2010-09-14 2014-02-19 Univ Kyoto Method of efficiently establishing induced pluripotent stem cells
CA2813466A1 (en) 2010-10-01 2012-04-05 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids that disrupt major groove binding partner interactions
WO2012048213A1 (en) 2010-10-08 2012-04-12 Regents Of The University Of Minnesota A method to increase gene targeting frequency
US9637732B2 (en) 2010-11-04 2017-05-02 Kyoto University Method of efficiently establishing induced pluripotent stem cells
US9267123B2 (en) 2011-01-05 2016-02-23 Sangamo Biosciences, Inc. Methods and compositions for gene correction
AU2012225497A1 (en) 2011-03-07 2013-10-24 Massachusetts Institute Of Technology Methods for transfecting cells with nucleic acids
WO2012131090A1 (en) 2011-03-31 2012-10-04 Galderma Research & Development Method for treatment of xeroderma pigmentosum
CA2831613A1 (en) 2011-03-31 2012-10-04 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
WO2012138453A1 (en) 2011-04-03 2012-10-11 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
US9315788B2 (en) 2011-04-05 2016-04-19 Cellectis, S.A. Method for the generation of compact TALE-nucleases and uses thereof
WO2012174224A2 (en) 2011-06-17 2012-12-20 Calando Pharmaceuticals, Inc. Methods for administering nucleic acid-based therapeutics
WO2012176015A1 (en) 2011-06-24 2012-12-27 Leo Pharma A/S Methods for treating uv-damaged skin and scc tumors and for removing tattoos with topical ingenol mebutate
WO2013003475A1 (en) 2011-06-27 2013-01-03 Cellscript, Inc. Inhibition of innate immune response
EP2732029B1 (en) 2011-07-11 2019-01-16 FUJIFILM Cellular Dynamics, Inc. Methods for cell reprogramming and genome engineering
CN103842517A (en) 2011-08-03 2014-06-04 莲花组织修复公司 Collagen 7 and related methods
CN110511939A (en) 2011-10-03 2019-11-29 现代泰克斯公司 Nucleosides, nucleotide and nucleic acid of modification and application thereof
ES2867054T3 (en) 2011-10-11 2021-10-20 Inst Nat Sante Rech Med Exon skipping treatment for dystrophic epidermolysis bullosa
WO2013078199A2 (en) * 2011-11-23 2013-05-30 Children's Medical Center Corporation Methods for enhanced in vivo delivery of synthetic, modified rnas
RU2624139C2 (en) * 2011-12-05 2017-06-30 Фэктор Байосайенс Инк. Methods and formulations for cells transfection
US8497124B2 (en) 2011-12-05 2013-07-30 Factor Bioscience Inc. Methods and products for reprogramming cells to a less differentiated state
US20140343129A1 (en) * 2011-12-14 2014-11-20 Moderna Therapeutics, Inc. Modified nucleic acids, and acute care uses thereof
EP2791160B1 (en) 2011-12-16 2022-03-02 ModernaTX, Inc. Modified mrna compositions
US20130165504A1 (en) 2011-12-21 2013-06-27 modeRNA Therapeutics Methods of increasing the viability or longevity of an organ or organ explant
PL3421601T3 (en) 2011-12-30 2020-06-01 Cellscript, Llc Making and using in vitro-synthesized ssrna for introducing into mammalian cells to induce a biological or biochemical effect
EP2833923A4 (en) * 2012-04-02 2016-02-24 Moderna Therapeutics Inc Modified polynucleotides for the production of proteins
US20130274129A1 (en) 2012-04-04 2013-10-17 Geneart Ag Tal-effector assembly platform, customized services, kits and assays
US20150211004A1 (en) * 2012-04-20 2015-07-30 Agency For Science, Technology And Research Rnai-based therapies for cardiomyopathies, muscular dystrophies and laminopathies
WO2013163296A1 (en) 2012-04-24 2013-10-31 The Brigham And Women's Hospital, Inc. Generating pluripotent cells de novo
DK2850179T3 (en) 2012-05-13 2019-06-17 Allele Biotechnology & Pharmaceuticals Inc FEEDER CELL-FREE DERIVATION OF HUMAN-INDUCED PLURIPOTENT STAM CELLS WITH SYNTHETIC MESSENGER RNA
US10155929B2 (en) 2012-05-13 2018-12-18 Allele Biotechnology & Pharmaceuticals, Inc. Feeder-free derivation of human-induced pluripotent stem cells with synthetic messenger RNA
US10119150B2 (en) 2012-05-13 2018-11-06 Allele Biotechnology & Pharmaceuticals, Inc. Feeder-free Derivation of human-induced pluripotent stem cells with synthetic messenger RNA
US20140031295A1 (en) 2012-07-19 2014-01-30 Lotus Tissue Repair, Inc. Recombinant C7 and Methods of Use
JP6510416B2 (en) 2012-11-01 2019-05-08 ファクター バイオサイエンス インコーポレイテッド Methods and products for expressing proteins in cells
PL2959005T3 (en) 2013-02-22 2022-01-31 The Board Of Trustees Of The Leland Stanford Junior University Medical use relating to telomere extension
US20140242595A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
WO2014134412A1 (en) 2013-03-01 2014-09-04 Regents Of The University Of Minnesota Talen-based gene correction
WO2015038075A1 (en) 2013-09-16 2015-03-19 Agency For Science, Technology And Research Method
JP6912887B2 (en) 2013-12-12 2021-08-04 ライフ テクノロジーズ コーポレーション Membrane-permeable peptides for enhancing transfection and compositions and methods using them
US20150167017A1 (en) * 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
RU2714404C2 (en) * 2014-01-31 2020-02-14 Фэктор Байосайенс Инк. Methods and products for producing and delivering nucleic acids
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Chatterjee et al. (2011, J. Visualized Exp., Vol. 56, pgs. 1-4). (Year: 2011) *
Hynes et al. (2013, J. Dent. Res., Vol. 92(9), pgs. 833-839). (Year: 2013) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11904023B2 (en) 2016-08-17 2024-02-20 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof

Also Published As

Publication number Publication date
CN109477102A (en) 2019-03-15
EP3543339A1 (en) 2019-09-25
JP2022000470A (en) 2022-01-04
US20180256748A1 (en) 2018-09-13
US11241505B2 (en) 2022-02-08
JP2018506542A (en) 2018-03-08
CA2976376A1 (en) 2016-08-18
AU2022206783A1 (en) 2022-08-25
AU2016218977C1 (en) 2023-03-23
EP3256585A4 (en) 2018-08-15
WO2016131052A1 (en) 2016-08-18
JP7199809B2 (en) 2023-01-06
AU2016218977A1 (en) 2017-08-17
EP3256585A1 (en) 2017-12-20
AU2016218977B2 (en) 2022-07-21

Similar Documents

Publication Publication Date Title
US11904023B2 (en) Nucleic acid products and methods of administration thereof
US20200261599A1 (en) Nucleic acid products and methods of administration thereof
US20210024907A1 (en) Nucleic acid-based therapeutics
NZ791466A (en) Nucleic Acid Products And Methods Of Administration Thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: FACTOR BIOSCIENCE INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ANGEL, MATTHEW;ROHDE, CHRISTOPHER;REEL/FRAME:051943/0728

Effective date: 20160224

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED