CA2471363C - Albumin fusion proteins - Google Patents

Albumin fusion proteins Download PDF

Info

Publication number
CA2471363C
CA2471363C CA2471363A CA2471363A CA2471363C CA 2471363 C CA2471363 C CA 2471363C CA 2471363 A CA2471363 A CA 2471363A CA 2471363 A CA2471363 A CA 2471363A CA 2471363 C CA2471363 C CA 2471363C
Authority
CA
Canada
Prior art keywords
hsa
glp
seq
amino acids
psac35
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.)
Expired - Fee Related
Application number
CA2471363A
Other languages
French (fr)
Other versions
CA2471363A1 (en
Inventor
Craig A. Rosen
William A. Haseltine
Steven M. Ruben
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.)
Human Genome Sciences Inc
Original Assignee
Human Genome Sciences 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27586544&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2471363(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Human Genome Sciences Inc filed Critical Human Genome Sciences Inc
Priority to CA2841097A priority Critical patent/CA2841097A1/en
Publication of CA2471363A1 publication Critical patent/CA2471363A1/en
Application granted granted Critical
Publication of CA2471363C publication Critical patent/CA2471363C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • A61P25/10Antiepileptics; Anticonvulsants for petit-mal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/40Mineralocorticosteroids, e.g. aldosterone; Drugs increasing or potentiating the activity of mineralocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/12Antidiuretics, e.g. drugs for diabetes insipidus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4713Autoimmune diseases, e.g. Insulin-dependent diabetes mellitus, multiple sclerosis, rheumathoid arthritis, systemic lupus erythematosus; Autoantigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4723Cationic antimicrobial peptides, e.g. defensins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5406IL-4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/57563Vasoactive intestinal peptide [VIP]; Related peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/5759Products of obesity genes, e.g. leptin, obese (OB), tub, fat
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/585Calcitonins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/59Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g. HCG; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/60Growth-hormone releasing factors (GH-RF) (Somatoliberin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/62Insulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/635Parathyroid hormone (parathormone); Parathyroid hormone-related peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/65Insulin-like growth factors (Somatomedins), e.g. IGF-1, IGF-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/655Somatostatins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/665Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans derived from pro-opiomelanocortin, pro-enkephalin or pro-dynorphin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0006Oxidoreductases (1.) acting on CH-OH groups as donors (1.1)
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0008Oxidoreductases (1.) acting on the aldehyde or oxo group of donors (1.2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/335Glucagon; Glucagon-like peptide [GLP]; Exendin
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16171Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y101/00Oxidoreductases acting on the CH-OH group of donors (1.1)
    • C12Y101/01Oxidoreductases acting on the CH-OH group of donors (1.1) with NAD+ or NADP+ as acceptor (1.1.1)
    • C12Y101/01105Retinol dehydrogenase (1.1.1.105)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y102/00Oxidoreductases acting on the aldehyde or oxo group of donors (1.2)
    • C12Y102/01Oxidoreductases acting on the aldehyde or oxo group of donors (1.2) with NAD+ or NADP+ as acceptor (1.2.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/01Phosphotransferases with an alcohol group as acceptor (2.7.1)
    • C12Y207/01095Kanamycin kinase (2.7.1.95), i.e. neomycin-kanamycin phosphotransferase

Abstract

The present invention encompasses albumin fusion proteins. Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention and using these nucleic acids, vectors, and/or host cells. Additionally the present invention encompasses pharmaceutical compositions comprising albumin fusion proteins and methods of treating, preventing, or ameliorating diseases, disorders or conditions using albumin fusion proteins of the invention.

Description

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.

NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional volumes please contact the Canadian Patent Office.

Albumin Fusion Proteins BACKGROUND OF THE INVENTION
[0001] The invention relates generally to Therapeutic proteins (including, but not limited to, at least one polypeptide, antibody, peptide, or fragment and variant thereof) fused to albumin or fragments or variants of albumin. The invention encompasses polynucleotides encoding therapeutic albumin fusion proteins, therapeutic albumin fusion proteins, compositions, pharmaceutical compositions, formulations and kits. Host cells transformed with the polynucleotides encoding therapeutic albumin fusion proteins are also encompassed by the invention, as are methods of making the albumin fusion proteins of the invention using these polynucleotides, and/or host cells.
[0002]
Human serum albumin (HSA, or HA), a protein of 585 amino acids in its mature form (as shown in Figure 1 (SEQ ID NO:1038)), is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands. At present, HA for clinical use is produced by extraction from human blood. The production of recombinant HA (rHA) in microorganisms has been disclosed in EP 330 451 and EP 361 991.
[0003]
Therapeutic proteins in their native state or when recombinantly produced, such as interferons and growth hormones, are typically labile molecules exhibiting short shelf-lives, particularly when formulated in aqueous solutions. The instability in these molecules when formulated for administration dictates that many of the molecules must be lyophilized and refrigerated at all times during storage, thereby rendering the molecules difficult to transport and/or store.
Storage problems are particularly acute when pharmaceutical formulations must be stored and dispensed outside of the hospital environment.
[0004] Few practical solutions to the storage problems of labile protein molecules have been proposed. Accordingly, there is a need for stabilized, long lasting formulations of proteinaceous therapeutic molecules that are easily dispensed, preferably with a simple formulation requiring minimal post-storage manipulation.

SUMMARY OF THE INVENTION
An object of the present invention is to provide albumin fusion proteins. In accordance with an aspect of the present invention, there is provided an albumin fusion protein comprising a member selected from the group consisting of:
(a) a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:1038;
(b) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:1038, wherein said fragment or variant has albumin activity;
(c) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:1038, wherein said fragment or variant has albumin activity, and further wherein said albumin activity is the ability to prolong the shelf life of the Therapeutic protein:X compared to the shelf-life of the Therapeutic protein:X in an unfused state;
(d) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:1038, wherein said fragment or variant has albumin activity, and further wherein the fragment or variant comprises the amino acid sequence of amino acids 1-387 of SEQ ID NO:1038;
(e) a fragment or variant of a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:1038, wherein said fragment or variant has a biological activity of the Therapeutic protein:X;
(0 a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the N-terminus of albumin, or the N-terminus of the fragment or variant of albumin;
(g) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the C-terminus of albumin, or the C-terminus of the fragment or variant of albumin;
(h) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the N- terminus and C-terminus of albumin, or the N-terminus and the C-tenninus of the fragment or variant of albumin;
(i) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), which comprises a first Therapeutic protein:X, or fragment or variant thereof, and a second Therapeutic protein:X, or fragment or variant thereof, wherein said first Therapeutic protein:X, or fragment or variant thereof, is different from said second Therapeutic protein:X, or fragment or variant thereof;
a Therapeutic protein:X, or fragment or variant thereof, and albumin, or 'fragment or variant thereof, of (a) to (i), wherein the Therapeutic protein:X, or fragment or variant thereof, is separated from the albumin or the fragment or variant of albumin by a linker; and (k) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (j), wherein the albumin fusion protein has the following formula:
= R1-L-R2; R2-L-R1; or R1-L-R2-L-R1, and further wherein R1 is Therapeutic protein:X, or fragment or variant thereof, L is a peptide linker, and R2 is albumin comprising the amino acid sequence of SEQ ID
NO:1038 or a fragment or variant of albumin.
In accordance with another aspect of the invention, there is provided an albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof, inserted into an albumin, or fragment or variant thereof, comprising the amino acid sequence of SEQ
ID NO:1038 or fragment or variant thereof.
In accordance with another aspect of the invention, there is provided an albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof, inserted into an albumin, or fragment or variant thereof, comprising an amino acid sequence selected from the group consisting of:
(a) amino acids 54 to 61 of SEQ ID NO:1038;
(b) amino acids 76 to 89 of SEQ ID NO:1038;
(c) amino acids 92 to100 of SEQ ID NO:1038;
(d) amino acids 170 to 176 of SEQ ID NO:1038;
(e) amino acids 247 to 252 of SEQ ID NO:1038;
2a (0 amino acids 266 to 277 of SEQ ID NO:1038;
(g) amino acids 280 to 288 of SEQ ID NO:1038;
(h) amino acids 362 to 368 of SEQ ID NO:1038;
(i) amino acids 439 to 447 of SEQ ID NO:1038;
(i) amino acids 462 to 475 of SEQ NO:1038;
(k) amino acids 478 to 486 of SEQ ID NO:1038; and amino acids 560 to 566 of SEQ ID NO:1038.
In accordance with another aspect of the invention, there is provided a method of extending the shelf life of Therapeutic protein:X, or fragment or variant thereof, comprising the step of fusing the Therapeutic protein:X, or fragment or variant thereof, to albumin, or fragment or variant thereof, sufficient to extend the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, compared to the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
The present invention encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin. The present invention also encompasses polynucleotides co.mprising, or alternatively consisting of, nucleic acid molecules encoding a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin.
The present invention also encompasses polynucleotides, comprising, or alternatively consisting of, nucleic acid molecules encoding proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin, that is sufficient to prolong the shelf life of the Therapeutic protein, and/or stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo. Albumin fusion proteins encoded by a polynucleotide of the invention are also encompassed by the invention, as are host cells transformed with polynucleotides of the invention, and methods of making the albumin fusion proteins of the invention and using these polynucleotides of the invention, and/or host cells.
2b In a preferred aspect of the invention, albumin fusion proteins include, but are not limited to, those encoded by the polynucleotides described in Table 2.
The invention also encompasses pharmaceutical formulations comprising an albumin fusion protein of the invention and a pharmaceutically acceptable diluent or carrier.
Such formulations may be in a kit or container. Such kit or container may be packaged with instructions pertaining to the extended shelf life of the Therapeutic protein.
Such formulations may be used in methods of treating, preventing, ameliorating or diagnosing a disease or disease symptom in a patient, preferably a mammal, most preferably a human, comprising the step of administering the pharmaceutical formulation to the patient.
In other embodiments, the present invention encompasses methods of preventing, treating, or ameliorating a disease or disorder. In preferred embodiments, the present invention encompasses a method of treating a disease or disorder listed in the "Preferred Indication: Y" column of Table 1 comprising administering to a patient in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Therapeutic protein or portion corresponding to a Therapeutic 2c protein (or fragment or variant thereof) disclosed in the "Therapeutic Protein: X" column of Table 1 (in the same row as the disease or disorder to be treated is listed in the "Preferred Indication: Y" column of Table 1) in an amount effective to treat, prevent or ameliorate the disease or disorder.
[0009] In one embodiment, an albumin fusion protein described in Table 1 or 2 has extended shelf life.
[0010] In a second embodiment, an albumin fusion protein described in Table 1 or 2 is more stable than the corresponding unfused Therapeutic molecule described in Table 1.
[0011] The present invention further includes transgenic organisms modified to contain the nucleic acid molecules of the invention (including, but not limited to, the polynucleotides described in Tables 1 and 2), preferably modified to express an albumin fusion protein of the invention.
BRIEF DESCRIPTION OF THE FIGURES
[0012] Figure 1A-D shows the amino acid sequence of the mature form of human albumin (SEQ ID NO:1038) and a polynucleotide encoding it (SEQ ID NO:1037).
[0013] Figure 2 shows the restriction map of the pPPC0005 cloning vector ATCC
deposit PTA-3278.
[0014] Figure 3 shows the restriction map of the pSAC35 yeast S.
cerevisiae expression vector (Sleep et al., BioTeclanology 8:42 (1990)).
[0015] Figure 4 shows the effect of various dilutions of EPO albumin fusion proteins encoded by DNA comprised in Construct ID NOS. (hereinafter CID) 1966 and 1981 and recombinant human EPO on the proliferation of TF-1 cells (see Examples 8 and 9). Cells were washed 3X to remove GM-CSF and plated at 10,000 cells/well for 72 hours in the presence of 3-fold dilutions of CUD 1966 protein or CID 1981 protein.
Concentrations used were calculated based on the weight of Epo alone, not HSA plus Epo.
Recombinant human Epo (rhEpo) was used as the positive control and serially diluted 3 fold from 100 ng/ml to 0.01 ng/ml. Cells were exposed to 0.5 mCi/well of 3H-thymidine for an additional 18 hours.
(0) rhEpo; (V) HSA-Epo 1981; (e) Epo-HSA 1966.
[0016] Figure 5 is a dose response analysis and shows the effect of various doses of recombinant human EPO and EPO albumin fusion proteins encoded by DNA comprised in CD 1966 and 1981 on the percent change in hematocrit from day 0 to day 7 (see Examples 8 and 9). 48 eight-week old female DBA/2NHsd mice were divided into 12 groups of animals each. Recombinant human Epo (rhEpo) was administered subcutaneously at 0.5, 1.5, 4.5 and 12 1.1g/kg on days 0, 2, 4, and 6. Epo albumin fusion proteins made from constructs CD 1966 and CID 1981 were administered subcutaneously at 2, 6, 18, and 54 p,g/kg on days 0, 2, 4, and 6. The higher doses of the Epo albumin fusion proteins allows a rough equimolar comparison with recombinant human Epo (note that the weight of the fusions is about 4.35 times the weight of non-glycosylated Epo). On days 0 and 7 of the experiment, the animals were bled via a tail vein and the hematocrit was determined by centrifugation.
(U) rhEpo; (o) OD 1981; (A) CID 1966.
[0017] Figure 6A shows the effect of various subcutaneous administrations of Epo albumin fusion proteins encoded by DNA comprised in CD 1966 and 1997, respectively, on the percent change in hematocrit from day 0 to day 8 (see Examples 8 and 10).
*, p<0.005 compared to rhEpo as determined by Mann-Whitney nonparametric analysis (n=6).
[0018] Figure 6B shows the effect of subcutaneous administrations of Epo albumin fusion proteins encoded by DNA comprised in CD 1997 and 1966 on the percent change in hematocrit from day 0 to day 14 (see Examples 8 and 10). *, p<0.005 compared to rhEpo as determined by Mann-Whitney nonparametric analysis (n=6); **, p<0.05 compared to rhEpo as determined by Mann-Whitney nonparametric analysis (n=6).
[0019] Figure 7 shows the effect of various dilutions albumin fusion proteins encoded by DNA comprised in OD 1981 and 1997, respectively, on the proliferation of TF-1 cells (see Examples 9 and 10). Cells were washed 3X to remove GM-CSF and plated at 10,000 cells/well for 72 hours in the presence of 3-fold dilutions of Epo albumin fusion proteins encoded by CD 1981 or 1997. Equimolar amounts of rhEpo were used as a positive control (4.35 times less protein added since weight of non-glycosylated Epo is 20 kd, while Epo albumin fusion proteins are 87 kd). Cells were exposed to 0.5 laCi/well of 3H-thymidine for an additional 24 hours. (M) rhEpo Standard; (A) CID 1981 (CHO); (o) OD 1997 (NSO).
[0020] Figure 8 shows the effect of various doses of recombinant human EPO
(rhEpo) and EPO albumin fusion protein encoded by DNA comprised in construct 1997 (CID
1997) on the percent change in hematocrit from day 0 to day 8 (see Example 10). (A) =
rhEpo, (0) = OD 1997.
[0021] Figure 9 shows the effect of various dilutions of IL2 albumin fusion proteins encoded by DNA comprised in CID 1812 (see Example 15) on CTLL-2 proliferation.
1x104 cells/well were seeded in a 96-well plate in a final volume of 200 ul of complete medium containing the indicated amount of IL2 albumin fusion protein (CID 1812). All samples were run in triplicate. The cells were incubated for 40 hours at 37 C, then 20 ul of Alamar Blue was added and cells incubated for 8 hours. Absorbance at 530/590 was used as a measure of proliferation. EC50 = 0.386 0.021. (A) = OD 1812.
[0022] Figure 10 shows the effect of 1L2 albumin fusion protein encoded by DNA
comprised in CID 1812 on RENCA tumor growth at day 21 (see Example 15). BALB/c mice (n=10) were injected SC (midflank) with 105 RENCA cells. 10 days later mice received 2 cycles (Day 10 to Day 14 and Days 17-21) of daily (QD) injections of rIL2 (0.9 mg/kg), IL2 albumin fusion protein (CID 1812 protein; 0.6 mg/kg), or PBS (Placebo) or injections every other day (QOD) of OD 1812 protein (0.6 mg/kg). The tumor volume was determined on Day 21 after RENCA inoculation. The data are presented in scatter analysis (each dot representing single animal). Mean value of each group is depicted by horizontal line. *, p=0.0035 between placebo control and CID 1812 protein. The number in parentheses indicates number of mice alive over the total number of mice per group. (0) =
Placebo; (0) =
EL2; (A) = CID 1812 protein (QD); (El) = CID 1812 protein (QOD).
[0023] Figure 11 shows the effect of various dilutions of GCSF albumin fusion proteins encoded by DNA comprised in OD 1642 and 1643 on NFS-60 cell proliferation (see Examples 19 and 20). (01) = OD 1642; (A) = CID 1643; (0) = HSA.
[0024] Figure 12 shows the effect of recombinant human GCSF (Neupogen) and GCSF albumin fusion protein on total white blood cell count (see Example 19).
Total WBC
(103 cells/ul) on each day are presented as the group mean SEM. GCSF albumin fusion protein was administered sc at either 25 or 100 ug/kg every 4 days x 4 (Q4D) , or at 100 ug/kg every 7 days x 2 (Q7D). Data from Days 8 and 9 for GCSF albumin fusion protein 100 ug/kg Q7 are presented as Days 9 and 10, respectively, to facilitate Comparison with other groups. Controls were saline vehicle administered SC every 4 days x 4 (Vehicle Q4D), or Neupogen administered SC daily x 14 (Neupogen 5 ug/kg QD). The treatment period is considered Days 1-14, and the recovery period, Days 15-28.
[0025] Figure 13 shows the effect of various dilutions of IFNb albumin fusion proteins encoded by DNA comprised in OD 2011 and 2053 on SEAP activity in the ISRE-SEAP/293F reporter cells (see Example 25). Proteins were serially diluted from 5e-7 to 1 e-14 g/ml in DMEM/10% FBS and used to treat ISRE-SEAP/293F reporter cells. After hours supernatants were removed from reporter cells and assayed for SEAP
activity. IFNb albumin fusion protein was purified from three stable clones: 293F/#2011, CH0/#2011 and NSO/#2053. Mammalian derived IFNb, Avonex, came from Biogen and was reported to have a specific activity of 2.0e5 IU/ug.
[0026] Figure 14 illustrates the steady-state levels of insulin mRNA in INS-1 (832/13) cells after treatment with GLP-1 or GLP-1 albumin fusion protein encoded by construct ID
3070 (CID 3070 protein). Both GLP-1 and the CID 3070 protein stimulate transcription of the insulin gene in INS-1 cells. The first bar (black) represents the untreated cells. Bars 2-4 (white) represent cells treated with the indicated concentrations of GLP-1.
Bars 5-7 (gray) represent cells treated with the indicated concentrations of CID 3070 protein.
[0027] Figure 15 compares the anti-proliferative activity of IFN albumin fusion protein encoded by CID 3165 (CID 3165 protein) and recombinant IFNa (rIFNa) on Hs294T
melanoma cells. The cells were cultured with varying concentrations of either protein or rIFNa and proliferation was measured by BrdU incorporation after 3 days of culture. CID 3165 protein caused measurable inhibition of cell proliferation at concentrations above 10 ng/ml with 50% inhibition achieved at approximately 200 ng/ml. (I) =

protein, (11) = rIFNa.
[0028] Figure 16 shows the effect of various dilutions of IFNa albumin fusion proteins on SEAP activity in the ISRE-SEAP/293F reporter cells. One preparation of IFNa fused upstream of albumin (40) was tested, as well as two different preparations of IFNa fused downstream of albumin (A) and ( = ).
[0029] Figure 17 shows the effect of time and dose of IFNa albumin fusion protein encoded by DNA comprised in construct 2249 (CD 2249 protein) on the mRNA level of OAS (01) in treated monkeys (see Example 31). Per time point: first bar =
Vehicle control, 2' bar 30 ug/kg CID 2249 protein day 1 iv, third bar = 30 ug/kg CID 2249 protein day 1 Sc, 4th bar = 300 ug/kg CID 2249 protein day 1 sc, 5th bar = 40 ug/kg recombinant IFNa day 1, 3 and 5 sc.
[0030] Figure 18 shows the effect of various dilutions of insulin albumin fusion proteins encoded by DNA comprised in constructs 2250 and 2276 on glucose uptake in 3T3-Li adipocytes (see Examples 33 and 35).
[0031] Figure 19 shows the effect of various GCSF albumin fusion proteins, including those encoded by CID #1643 and #2702 (L-171, see Example 114), on NFS cell proliferation. The horizontal dashed line indicates the minimum level of detection.
DETAILED DESCRIPTION

Definitions [0032] The following definitions are provided to facilitate understanding of certain terms used throughout this specification.
[0033] As used herein, "polynucleotide" refers to a nucleic acid molecule having a nucleotide sequence encoding a fusion protein comprising, or alternatively consisting of, at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one Therapeutic protein X (or fragment or variant thereof); a nucleic acid molecule having a nucleotide sequence encoding a fusion protein comprising, or alternatively consisting of, the amino acid sequence of SEQ ID NO:Y (as described in column 6 of Table 2) or a fragment or variant thereof; a nucleic acid molecule having a nucleotide sequence comprising or alternatively consisting of the sequence shown in SEQ ID NO:X; a nucleic acid molecule having a nucleotide sequence encoding a fusion protein comprising, or alternatively consisting of, the amino acid sequence of SEQ ID NO:Z; a nucleic acid molecule having a nucleotide sequence encoding an albumin fusion protein of the invention generated as described in Table 2 or in the Examples; a nucleic acid molecule having a nucleotide sequence encoding a Therapeutic albumin fusion protein of the invention, a nucleic acid molecule having a nucleotide sequence contained in an albumin fusion construct described in Table 2, or a nucleic acid molecule having a nucleotide sequence contained in an albumin fusion construct deposited with the ATCC (as described in Table 3).
[0034] As used herein, "albumin fusion construct" refers to a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof); a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof) generated as described in Table 2 or in the Examples; or a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least .one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof), further comprising, for example, one or more of the following elements: (1) a functional self-replicating vector (including but not limited to, a shuttle vector, an expression vector, an integration vector, and/or a replication system), (2) a region for initiation of transcription (e.g., a promoter region, such as for example, a regulatable or inducible promoter, a constitutive promoter), (3) a region for termination of transcription, (4) a leader sequence, and (5) a selectable marker. The polynucleotide encoding the Therapeutic protein and albumin protein, once part of the albumin fusion construct, may each be referred to as a "portion," "region" or "moiety" of the albumin fusion construct.
[0035] The present invention relates generally to polynucleotides encoding albumin fusion proteins; albumin fusion proteins; and methods of treating, preventing, or ameliorating diseases or disorders using albumin fusion proteins or polynucleotides encoding albumin fusion proteins. As used herein, "albumin fusion protein" refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof). An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin). The Therapeutic protein and albumin protein, once part of the albumin fusion protein, may each be referred to as a "portion", "region" or "moiety" of the albumin fusion protein (e.g., a "Therapeutic protein portion" or an "albumin protein portion"). In a highly preferred embodiment, an albumin fusion protein of the invention comprises at least one molecule of a Therapeutic protein X or fragment or variant of thereof (including, but not limited to a mature form of the Therapeutic protein X) and at least one molecule of albumin or fragment or variant thereof (including but not limited to a mature form of albumin).
[0036] In a further preferred embodiment, an albumin fusion protein of the invention is processed by a host cell and secreted into the surrounding culture medium.
Processing of the nascent albumin fusion protein that occurs in the secretory pathways of the host used for expression may include, but is not limited to signal peptide cleavage;
formation of disulfide bonds; proper folding; addition and processing of carbohydrates (such as for example, N- and 0- linked glycosylation); specific proteolytic cleavages; and assembly into multimeric proteins. An albumin fusion protein of the invention is preferably in the processed form. In a most preferred embodiment, the "processed form of an albumin fusion protein"
refers to an albumin fusion protein product which has undergone N- terminal signal peptide cleavage, herein also referred to as a "mature albumin fusion protein".

[0037] In several instances, a representative clone containing an albumin fusion construct of the invention was deposited with the American Type Culture Collection (herein referred to as "ATCCO"). Furthermore, it is possible to retrieve a given albumin fusion construct from the deposit by techniques known in the art and described elsewhere herein.
The ATCCO is located at 10801 University Boulevard, Manassas, Virginia 20110-2209, USA. The ATCCS deposits were made pursuant to the terms of the Budapest Treaty on the international recognition of the deposit of microorganisms for the purposes of patent procedure.
[0038] In one embodiment, the invention provides a polynucleotide encoding an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein. In a further embodiment, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein. In a preferred embodiment, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein encoded by a polynucleotide described in Table 2. In a further preferred embodiment, the invention provides a polynucleotide encoding an albumin fusion protein whose sequence is shown as SEQ ID NO:Y in Table 2. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein. In preferred embodiments, the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin. The invention further encompasses polynucleotides encoding these albumin fusion proteins.
[0039] In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin. In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin. In preferred embodiments, the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein. In a further preferred embodiment, the Therapeutic protein portion of the albumin fusion protein is the extracellular soluble domain of the Therapeutic protein. In an alternative embodiment, the Therapeutic protein portion of the albumin fusion protein is the active form of the Therapeutic protein. The invention further encompasses polynucleotides encoding these albumin fusion proteins.
[0040] In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin. In preferred embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin. The invention further encompasses polynucleotides encoding these albumin fusion proteins.
Therapeutic proteins [0041] As stated above, a polynucleotide of the invention encodes a protein comprising or alternatively consisting of, at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion.
[0042] An additional embodiment includes a polynucleotide encoding a protein comprising or alternatively consisting of at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are linked with one another by chemical conjugation.
[0043] As used herein, "Therapeutic protein" refers to proteins, polypeptides, antibodies, peptides or fragments or variants thereof, having one or more therapeutic and/or biological activities. Therapeutic proteins encompassed by the invention include but are not limited to, proteins, polypeptides, peptides, antibodies, and biologics. (The terms peptides, proteins, and polypeptides are used interchangeably herein.) It is specifically contemplated that the term "Therapeutic protein" encompasses antibodies and fragments and variants thereof. Thus a protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an antibody.
Additionally, the term "Therapeutic protein" may refer to the endogenous or naturally occurring correlate of a Therapeutic protein.
[0044] By a polypeptide displaying a "therapeutic activity" or a protein that is "therapeutically active" is meant a polypeptide that possesses one or more known biological and/or therapeutic activities associated with a therapeutic protein such as one or more of the Therapeutic proteins described herein or otherwise known in the art. As a non-limiting example, a "Therapeutic protein" is a protein that is useful to treat, prevent or ameliorate a disease, condition or disorder. As a non-limiting example, a "Therapeutic protein" may be one that binds specifically to a particular cell type (normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)) and therefore may be used to target a compound (drug, or cytotoxic agent) to that cell type specifically.
[0045] For example, a non-exhaustive list of "Therapeutic protein"
portions which may be comprised by an albumin fusion protein of the invention includes, but is not limited to, erythropoietin (EPO), IL-2, G-CSF, Insulin, Calcitonin, Growth Hormone, LFN-alpha, IFN-beta, PTH, TR6 (International Publication No. WO 98/30694), BLyS, BLyS
single chain antibody, Resistin, Growth hormone releasing factor, VEGF-2, KGF-2, D-SLAM, KDL and TR2, GLP-1, Extendin 4, and GM-CSF.
[0046] Interferon hybrids may also be fused to the amino or carboxy terminus of albumin to folin an interferon hybrid albumin fusion protein. Interferon hybrid albumin fusion protein may have enhanced, or alternatively, suppressed interferon activity, such as antiviral responses, regulation of cell growth, and modulation of immune response (Lebleu et al., PNAS USA, 73:3107-3111 (1976); Gresser et al., Nature, 251:543-545 (1974); and Johnson, Texas Reports Biol Med, 35:357-369 (1977)). Each interferon hybrid albumin fusion protein can be used to treat, prevent, or ameliorate viral infections (e.g., hepatitis (e.g., HCV); or HIV), multiple sclerosis, or cancer.
[0047] In one embodiment, the interferon hybrid portion of the interferon hybrid albumin fusion protein comprises an interferon alpha-interferon alpha hybrid (herein referred to as an alpha-alpha hybrid). For example, the alpha-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to interferon alpha D. In a further embodiment, the AID hybrid is fused at the common BgBI
restriction site to interferon alpha D, wherein the N-terminal portion of the A/D hybrid corresponds to amino acids 1-62 of interferon alpha A and the C-terminal portion corresponds to amino acids 64-166 of interferon alpha D. For example, this AID
hybrid would comprise the amino acid sequence:

SILAVKKYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRKE (SEQ ID
NO:1326), wherein the X1 is R or K and the X2 is A or V (see, for example, Construct ID
#2875). In an additional embodiment, the AID hybrid is fused at the common Pvu.ITI

restriction site, wherein the N-terminal portion of the AJD hybrid corresponds to amino acids 1-91 of interferon alpha A and the C-terminal portion corresponds to amino acids 93-166 of interferon alpha D. For example, this A/D hybrid would comprise the amino acid sequence:
CDLP QTHSLGSRRTLMLLAQMRXIIS IFS CLKDRHDFGFPQEEFGNQFQKAETIPVLHE

SILAVKKYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRKE (SEQ ID
NO:1311), wherein the X1 is R or K and the second X2 is A or V (see, for example, Construct ED #2872). These hybrids are further described in U.S. Patent No. 4,414,510.
[00481 In an additional embodiment, the alpha-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to interferon alpha F. In a further embodiment, the A/F hybrid is fused at the common PvullI
restriction site, wherein the N-terminal portion of the A/F hybrid corresponds to amino acids 1-91 of interferon alpha A and the C-terminal portion corresponds to amino acids 93-166 of interferon alpha F. For example, this .AJF hybrid would comprise the amino acid sequence:
CDLPQTHSLGSRRTLMLLAQMRXISLFS CLKDRHDFGFPQEEFGNQFQKAETIPVLHE
MIQQIENLFSTKD S S AAWDETLLDKFYTELYQQLNDMEACVIQEVGVEETPLMNVD S
ILAVKKYFQRITLYLTEKKYSPCAWEVVRAELVMSFSLSKIFQERLRRKE (SEQ ID
NO:1321), wherein X is either R or K (see, for example, Construct ID #2874).
These hybrids are further described in U.S. Patent No. 4,414,510.
In a further embodiment, the alpha-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to interferon alpha B. In an additional embodiment, the A/B hybrid is fused at the common Pvulll restriction site, wherein the N-terminal portion of the A/B hybrid corresponds to amino acids 1-91 of interferon alpha A and the C-terminal portion corresponds to amino acids 93-166 of interferon alpha B. For example, this A/B hybrid would comprise an amino acid sequence:
CDLPQTHSLGSRRTLMLLAQMRXIISLFSCLKDRHDFGFPQEEFONQFQKAETIPVLHE
MIQQIFNLFSTKDSSAAWDETLI .DKFYTELYQQLNDLEX2X3X4X5QEVGVIESPLMYE
DSILAVRKYFQRITLYLTEKKYSSCAWEVVRABIMRSFSLSINLQKRLKSKE (SEQ ID
NO:1316), wherein the X1 is R or K and X2 through X5 is SCVM or VLCD (see, for example, Construct ID #2873). These hybrids are further described in U.S. Patent No.
4,414,510.

[0049) In another embodiment, the interferon hybrid portion of the interferon hybrid albumin fusion protein comprises an interferon beta-interferon alpha hybrid (herein referred to as a beta-alpha hybrid). For example, the beta-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon beta-1 fused to interferon alpha D (also referred to as interferon alpha-1). In a further embodiment, the beta-1/alpha D hybrid is fused wherein the N-terminal portion corresponds to amino acids 1-73 of interferon beta-I and the C-terminal portion corresponds to amino acids 74-167 of interferon alpha D. For example, this beta-1/alpha D hybrid would comprise an amino acid sequence:
MSYNLLGFLQRSSNFQCQKLLWQLNGRLEYCLKDRMNFDIPEEIK.QLQQFQKEDAAL

XDSILAVKICYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRKE (SEQ
ID NO:2130), wherein X is A or V. These hybrids are further described in U.S.
Patent No.
4,758,428.
[0050] In another embodiment, the interferon hybrid portion of the interferon hybrid albumin fusion protein comprises an interferon alpha-interferon beta hybrid (herein referred to as a alpha-beta hybrid). For example, the alpha-beta hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha D (also referred to as interferon alpha-1) fused to interferon beta-1. In a further embodiment, the alpha D/beta-1 hybrid is fused wherein the N-terminal portion corresponds to amino acids 1-73 of interferon alpha D and the C-terminal portion corresponds to amino acids 74-166 of interferon beta-1. For example, this alpha D/beta-1 hybrid would have an amino acid sequence:
MCDLPETHSLDNRRTLMLLAQMSRISPSS CLMDRHDFGFPQEEFDGNQFQKAPAISV

MS SIELKRYYGRILHYLKAKEYSHCAWTIVRVEMRNFYFINRLTGYLRN (SEQ ID
NO:2131). These hybrids are further described in U.S. Patent No. 4,758,428_ [0051] In further embodiments, the interferon hybrid portion of the interferon hybrid albumin fusion proteins may comprise additional combinations of alpha-alpha interferon hybrids, alpha-beta interferon hybrids, and beta-alpha interferon hybrids. In additional embodiments, the interferon hybrid portion of the interferon hybrid albumin fusion protein may be modified to include mutations, substitutions, deletions, or additions to the amino acid sequence of the interferon hybrid. Such modifications to the interferon hybrid albumin fusion proteins may be made, for example, to improve levels of production, increase stability, increase or decrease activity, or confer new biological properties.
[0052] The above-described interferon hybrid albumin fusion proteins are encompassed by the invention, as are host cells and vectors containing polynucleotides encoding the polypeptides. In one embodiment, a interferon hybrid albumin fusion protein encoded by a polynucleotide as described above has extended shelf life. In an additional embodiment, a interferon hybrid albumin fusion protein encoded by a polynucleotide described above has a longer serum half-life and/or more stabilized activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo than the corresponding unfused interferon hybrid molecule.
[0053] In another non-limiting example, a "Therapeutic protein" is a protein that has a biological activity, and in particular, a biological activity that is useful for treating, preventing or ameliorating a disease. A non-inclusive list of biological activities that may be possessed by a Therapeutic protein includes, enhancing the immune response, promoting angiogenesis, inhibiting angiogenesis, regulating endocrine function, regulating hematopoietic functions, stimulating nerve growth, enhancing an immune response, inhibiting an immune response, or any one or more of the biological activities described in the "Biological Activities" section below and/or as disclosed for a given Therapeutic protein in Table 1 (column 2).
[0054] As used herein, "therapeutic activity" or "activity" may refer to an activity whose effect is consistent with a desirable therapeutic outcome in humans, or to desired effects in non-human mammals or in other species or organisms. Therapeutic activity may be measured in vivo or in vitro. For example, a desirable effect may be assayed in cell culture.
As an example, when EPO is the Therapeutic protein, the effects of EPO on cell proliferation as described in Example 8 may be used as the endpoint for which therapeutic activity is measured. Such in vitro or cell culture assays are commonly available for many Therapeutic proteins as described in the art. Examples of assays include, but are not limited to those described herein in the Examples section or in the "Exemplary Activity Assay"
column (column 3) of Table 1.
[0055] Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, such as cell surface and secretory proteins, are often modified by the attachment of one or more oligosaccharide groups. The modification, referred to as glycosylation, can dramatically affect the physical properties of proteins and can be important in protein stability, secretion, and localization. Glycosylation occurs at specific locations along the polypeptide backbone. There are usually two major types of glycosylation:
glycosylation characterized by 0-linked oligosaccharides, which are attached to serine or threonine residues; and glycosylation characterized by N-linked oligosaccharides, which are attached to asparagine residues in an Asn-X-Ser or Asn-X-Thr sequence, where X
can be any amino acid except proline. N-acetylneuramic acid (also known as sialic acid) is usually the terminal residue of both N-linked and 0-linked oligosaccharides. Variables such as protein structure and cell type influence the number and nature of the carbohydrate units within the chains at different glycosylation sites. Glycosylation isomers are also common at the same site within a given cell type.
[0056] For example, several types of human interferon are glycosylated. Natural human interferon-a2 is 0-glycosylated at threonine 106, and N-glycosylation occurs at asparagine 72 in interferon-a14 (Adolf et al., J. Biochem 276:511 (1991);
Nyman TA et al., J. Biochem 329:295 (1998)). The oligosaccharides at asparagine 80 in natural interferon-131a may play an important factor in the solubility and stability of the protein, but may not be essential for its biological activity. This permits the production of an unglycosylated analog (interferon-nib) engineered with sequence modifications to enhance stability (Hosoi et al., J.
Interferon Res. 8:375 (1988; Karpusas et al., Cell Mol Life Sci 54:1203 (1998); Knight, J.
Interferon Res. 2:421 (1982); Runkel et al., Pharm Res 15:641 (1998); Lin, Dev. Biol. Stand.
96:97 (1998)). Interferon-'y contains two N-linked oligosaccharide chains at positions 25 and 97, both important for the efficient formation of the bioactive recombinant protein, and having an influence on the pharmacokinetic properties of the protein (Sareneva et al., Fur. J.
Biochem 242:191 (1996); Sareneva et al,. Biochem J. 303:831 (1994); Sareneva et al., J.
Interferon Res. 13:267 (1993)). Mixed 0-linked and N-linked glycosylation also occurs, for example in human erythropoietin, N-linked glycosylation occurs at asparagine residues located at positions 24, 38 and 83 while 0-linked glycosylation occurs at a serine residue located at position 126 (Lai et al., J. Biol. Chem. 261:3116 (1986); Broudy et al., Arch.
Biochem. Biophys. 265:329 (1988)).
[0057]
Glycosylation of EPO albumin fusion proteins may influence the activity and/or stability of the EPO albumin fusion proteins. The EPO portion of the albumin fusion protein may contain 3 N-linked sites for glycosylation, each of which can carry one tetra-antennary structure. When the EPO albumin fusion protein is glycosylated, the half-life of the molecule may be increased. In one embodiment, the EPO albumin fusion protein is glycosylated. In another embodiment, the EPO albumin fusion protein is hyperglycosylated.
[0058] One type of sugar commonly found in oligosaccharides is sialic acid. Each tetra-antennary structure of the N-linked glycosylation sites of EPO may carry four sialic acid residues. Accordingly, in a preferred embodiment, the EPO albumin fusion protein is glycosylated with a carbohydrate group containing sialic acid. In an additional embodiment, the EPO albumin fusion protein comprises a fully sialylated EPO protein containing four sialic acid residues per tetra-antennerary structure per site with a molar ratio of sialic acid to protein 12:1 or greater. In alternative embodiments, the EPO albumin fusion protein comprises a hypersialylated EPO protein wherein one, two, or three sialic acid residues are attached at each tetra-antennerary structure per site with a molar ratio of sialic acid to protein less than 12:1.
[0059] Two types of sialic acid that may be used in the sialylation of the EPO
albumin fusion protein are N-acetylneurarninic acid (Neu5Ac) or N-glycolylneuraminic acid (Neu5Gc). In a preferred embodiment, hypersialylated EPO albumin fusion proteins contain Neu5Ac. More preferably, the total sialic acid content of hypersialylated EPO
albumin fusion proteins is at least 97% Neu5Ac. Most preferred are EPO albumin fusion protein structures with little or no Neu5Gc.
[0060] Preferably, the albumin EPO fusion protein has at least 4 moles of sialylation, and more preferably, at least 8-9 moles of sialylation. An additional embodiment comprises an albumin EPO fusion protein with 4 moles of sialylation, 5 moles of sialylation, 6 moles of sialylation, 7 moles of sialylation, 8-9 moles of sialylation, 8 moles of sialylation, 9 moles of sialylation, 10 moles of sialylation, 11 moles of sialylation, or 12 moles of sialylation.
[0061] The degree of sialylation of a protein changes the charge of the protein and its retention time on a chromatography column. Therefore, certain chromatography steps used in the purification process may be used to monitor or enrich for hypersialylated EPO albumin fusion proteins. In a preferred embodiment, the amount of sialylation may be monitored by HPLC chromatography. In an additional embodiment, steps in the purification process of EPO albumin fusions may be used to enrich for hypersialylated EF'0 albumin fusion proteins.
In a preferred embodiment the purification steps that may be used to enrich for hypersialylated EPO albumin fusion proteins comprise the butyl-sepharosTemFF
purification step to remove virus particles by high ammonium salt and the hydroxyapatite chromatography at pH 6.8 for the final purification step.
10062] Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, as well as analogs and variants thereof, may be modified so that glycosylation at one or more sites is altered as a result of manipulation(s) of their nucleic acid sequence, by the host cell in which they are expressed, or due to other conditions of their expression. For example, glycosylation isomers may be produced by abolishing or introducing glycosylation sites, e.g., by substitution or deletion of amino acid residues, such as substitution of glutamine for asparagine, or unglycosylated recombinant proteins may be produced by expressing the proteins in host cells that will not glycosylate them, e.g. in E. coli or glycosylation-deficient yeast. These approaches are described in more detail below and are known in the art.
[0063] Therapeutic proteins, particularly those disclosed in Table 1, and their nucleic acid and amino acid sequences are well known in the art and available in public databases such as Chemical Abstracts Services Databases (e.g., the CAS Registry), GenBank, and subscription provided databases such as GenSeq (e.g., Derwent). Exemplary nucleotide sequences of Therapeutic proteins which may be used to derive a polynucleotide of the invention are shown in column 7, "SEQ ID NO:X," of Table 2. Sequences shown as SEQ ID
NO:X may be a wild type polynucleotide sequence encoding a given Therapeutic protein (e.g., either full length or mature), or in some instances the sequence may be a variant of said wild type polynucleotide sequence (e.g., a polynucleotide which encodes the wild type Therapeutic protein, wherein the DNA sequence of said polynucleotide has been optimized, for example, for expression in a particular species; or a polynucleotide encoding a variant of the wild type Therapeutic protein (i.e., a site directed mutant; an allelic variant)). It is well ' within the ability of the skilled artisan to use the sequence shown as SEQ ID
NO:X to derive the construct described in the same row. For example, if SEQ ID NO:X
corresponds to a full length protein, but only a portion of that protein is used to generate the specific OD, it is within the skill of the art to rely on molecular biology techniques, such as PCR, to amplify the specific fragment and clone it into the appropriate vector.
[0064] Additional Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, one or more of the Therapeutic proteins or peptides disclosed in the "Therapeutic Protein X" column of Table 1 (column 1), or fragment or variable thereof.
[0065] Table 1 provides a non-exhaustive list of Therapeutic proteins that correspond to a Therapeutic protein portion of an albumin fusion protein of the invention, or an albumin fusion protein encoded by a polynucleotide of the invention. The first column, "Therapeutic Protein X," discloses Therapeutic protein molecules that may be followed by parentheses containing scientific and brand names of proteins that comprise, or alternatively consist of, that Therapeutic protein molecule or a fragment or variant thereof.
"Therapeutic protein X"
as used herein may refer either to an individual Therapeutic protein molecule, or to the entire group of Therapeutic proteins associated with a given Therapeutic protein molecule disclosed in this column. The "Biological activity" column (column 2) describes Biological activities associated with the Therapeutic protein molecule. Column 3, "Exemplary Activity Assay,"
provides references that describe assays which may be used to test the therapeutic and/or biological activity of a Therapeutic protein:X or an albumin fusion protein comprising a Therapeutic protein X (or fragment thereof) portion.
The fourth column, "Preferred Indication: Y," describes disease, disorders, and/or conditions that may be treated, prevented, diagnosed, and/or ameliorated by Therapeutic protein X or an albumin fusion protein comprising a Therapeutic protein X (or fragment thereof) portion. The "Construct ID" column (column 5) provides a link to an exemplary albumin fusion construct disclosed in Table 2 which encodes an albumin fusion protein comprising, or alternatively consisting of the referenced Therapeutic Protein X (or fragment thereof) portion.

Table 1 c Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
_ Construct ID Therapeutic Protein:X Assay Protein:Z 'a EPO Stimulates cellular Cell proliferation assay Anemia; Anemia in Renal Disease; Anemia in 1772, 1774, 1781, 1783, See Table 2, =

. (Erythropoietin; differentiation of bone- using a erythroleukemic cell Oncology Patients; Bleeding Disorders; Chronic 1793, 1794, 1925, 1926, SEQ
ID NO:Z 1--, Epoetin alfa; marrow stem cells at an line TF-1. (Kitamura et al. Renal Failure; Chronic Renal Failure in Pre- 1966, 1969, 1980, 1981, for particular Epoetin beta; early stage of 1989 J.Cell. Physiol.
Dialysis Patients; Renal Disease; End-Stage 1994, 1995, 1996, 1997, construct.
Gene-activated erythropoiesis; 140:323) Renal Disease; End-Stage Renal Disease in 2047, 2102, 2283, 2284, erythropoietin; accelerates the Dialysis Patients;
Chemotherapy; Chemotherapy 2287, 2289, 2294, 2298, Darbepoetin- proliferation and in Cancer Patients;
Anemia in zidovudine-treated 2310, 2311, 2325, 2326, alpha; NESP; maturation of terminally HIV patients; Anemia in zidovudine-treated 2344, 2363, 2373, 2387, Epogen; Procrit; differentiating cells into patients; Anemia in HIV patients; Anemia in 2414, 2441, 2603, 2604, n Eprex; Erypo; erythrocytes; and premature infants;
Surgical patients (pre and/or 2605, 3194, 3195, 3196, Espo; Epoimmun; modulates the level of post surgery);
Surgical patients (pre and/or post 0 I.) EPOGIN; circulating erythrocytes. surgery) who are anemic; Surgical patients (pre a, -.3 NEORECORMO and/or post surgery) who are undergoing elective H
CA
c7, N; HEMOLINK; surgery; Surgical patients (pre and/or post u.) Dynepo; surgery) who are undergoing elective, non- N) ARANESP) cardiac surgery;
Surgical patients (pre and/or 0 a, I
post surgery) who are undergoing elective, non-c7, cardiac, non-vascular surgery; Surgical patients I.) (pre and/or post surgery) who are undergoing H
elective, non-vascular surgery; Surgical patients (pre and/or post surgery) who are undergoing cardiac and/or vascular surgery; Aplastic anemia;
Refractory anemia; Anemia in Inflammatory Bowel Disease; Refractory anemia in Inflammatory Bowel Disease; Transfusion avoidance; Transfusion avoidance for surgical Iv patients; Transfusion avoidance for elective n surgical patients; Transfusion avoidance for elective orthopedic surgical patients; Patients cp who want to Increase Red Blood Cells.
G-CSF Stimulates the Proliferation of murine Chemoprotection; Adjunct to Chemotherapy; 1642, 1643, 2363, 2373, See Table 2, a (Granulocyte proliferation and NES-60 cells (Weinstein et Inflammatory disorders; Cancer; Leukemia; 2387, 2414, 2441, 2702, SEQ ID
NO:Z vD
1--, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a -a-, Protein:X Assay Protein:Z o o stimulating factor; progenitor cells for A 1986; 83, pp5010-4) neutropenias (e.g.; Kostmann syndrome); 2886, 2887, 2888, 2889, construct. =
--.1 Granulokine; granulocytes and Secondary neutropenia;
Prevention of 2890, 1--, KRN 8601; monocytes-macrophages. neutropenia; Prevention and treatment of Filgrastim; neutropenia in HIV-infected patients; Prevention Lenograstim; and treatment of neutropenia associated with Meograstim; chemotherapy; Infections associated with Nartograstim; neutropenias;
Myelopysplasia; Autoimmune Neupogen; disorders; Psoriasis;
Mobilization of NOPIA; Gran; hematopoietic progenitor cells; Wound Healing;
n GRANOCYTE; Autoimmune Disease;
Transplants; Bone marrow Granulolcine; transplants; Acute myelogeneous leukemia; 0 I.) Neutrogin; Neu- Lymphoma, Non-Hodgkin's lymphoma; Acute a, -.3 H
up; Neutromax) lymphoblastic leukemia;
Hodgkin's disease; u.) c7, Accelerated myeloid recovery; Glycogen storage u.) t\.) disease.
I.) c) GM-CSF Regulates hematopoietic Colony Stimulating Assay: Bone Marrow Disorders; Bone marrow 1697, 1699, 2066, and 2067. See Table 2, 0 a, (Granulocyte- cell differentiation, gene Testa, N.G., et al., "Assays transplant; Chemoprotection; Hepatitis C; HIV SEQ ID NO:Z 0 c7, I.) colony- function. factors." Balkwill FR (edt) Malignant melanoma; Mycobacterium avium construct. H
stimulating factor; Cytokines, A practical complex; Mycoses;
Leukemia; Myeloid rhuGM-CSF; BI Approach, pp 229-44; IRL Leukemia;
Infections; Neonatal infections;
61012; Prokine; Press Oxford 1991. Neutropenia;
Mucositis; Oral Mucositis; Prostate Molgramostim; Cancer; SternCell Mobilization; Vaccine Sargramostim; Adjuvant; Ulcers (such as Diabetic, Venous GM-CSF/IL 3 Stasis, or Pressure Ulcers); Prevention of fusion; neutropenia; Acute myelogenous leukemia; Iv Milodistim; Hematopoietic progenitor cell mobilization; n ,-i Leucotropin; Lymphoma; Non-Hodgkin's lymphoma; Acute PROKINE; Lymphoblastic Leukemia;
Hodgkin's disease; cp o LEUKOMAX; Accelerated myeloid recovery; Transplant n.) .-1,..-Interberin; Rejection;
Xenotransplant Rejection. a Leukine; Leulcine o 1--, Liquid; Pixykine) !

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a -a 5 Protein:X Assay Protein:Z o o Human growth Binds to two GHR Ba/F3-hGHR proliferation Acromegaly; Growth failure; Growth hormone 3163, 2983, See Table 2, =

hormone molecules and Induces assay, a novel specific replacement; Growth hormone deficiency; SEQ ID
NO:Z 1--, (Pegvisamont; signal transduction bioassay for serum human Pediatric Growth Hormone Deficiency; Adult for particular Somatrem; through receptor growth hormone. J Clin Growth Hormone Deficiency; Idiopathic Growth construct.
Somatropin; dimerization Endocrinol Metab 2000 Hormone Deficiency;
Growth retardation;
TROVERT; Nov;85(11):4274-9 Prader-Willi Syndrome;
Prader-Willi Syndrome PROTROPIN; Plasma growth hormone in children 2 years or older; Growth deficiencies;
BIO-TROPIN; (GH) immunoassay and Growth failure associated with chronic renal HUMATROPE; tibial bioassay, Appl insufficiency;
Osteoporosis; Postmenopausal n NUTROPIN; Physiol 2000 osteoporosis;
Osteopenia, Osteoclastogenesis;
NUTROPIN AQ; Dec;89(6):2174-8 burns; Cachexia; Cancer Cachexia; Dwarfism; 2 NUTROPH1N; Growth hormone (hGH) Metabolic Disorders;
Obesity; Renal failure; ti NORDITROP1N; receptor mediated cell Turner's Syndrome; Fibromyalgia;
Fracture ro c7, GENOTROPIN; mediated proliferation, treatment; Frailty, AIDS
wasting; Muscle u.) I") SAIZEN; Growth Hoim.IGF Res 2000 Wasting; Short Stature; Diagnostic Agents; 10) SEROSTIM) Oct;10(5):248-55 Female Infertility;
lipodystrophy. 0 a, International standard for c7, growth hormone, Horm Res iv 1999;51 Suppl 1:7-12 H
Insulin (Human Stimulates glucose uptake Insulin activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; 2250, 2255, 2276, 2278, See Table 2, Glargine; Insulin lipogenesis. Biol Chem 1999 Oct 22;
Hyperlipidemia; Hyperketonemia; Non-insulin 2878, 2882, 2885, 2891, construct.
lispro; Lys-B28 274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin- 2897, 2930, 2931, 2942, Pro-B29; lyspro; dependent Diabetes Mellitus (IDDM); A 2986, 3025, 3133, 3134, LY 275585; Condition Associated With Diabetes Including, 3197, 3198, 2726, 2727, Iv diarginylinsulin; But Not Limited To Obesity, Heart Disease, 2784, 2789 n Des-B26-B30- Hyperglycemia, Infections, Retinopathy, And/Or 1-3 insulin-B25- Ulcers; Metabolic Disorders; Immune Disorders; cp amide; Insulin Obesity; Vascular Disorders; Suppression of detemir; LABI; Body Weight; Suppression of Appetite; a NOVOLIN; Syndrome X.
'4 NOVORAPID;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z 'a o HUMULIN;
=
o NOVOMIX 30;

1--, VELOSULIN;
NOVOLOG;
LANTUS;
ILETIN; -HUMALOG;
MACRULIN;
EXUBRA;
INSUMAN;
n ORALIN;

ORALGEN;
I.) a,.
HUMAHALE;
H
CA
HUMAHALIN) m us, L-3 Interferon alfa Confers a range of Anti-viral assay: Rubinstein Viral infections; HIV Infections;
Hepatitis; 2249, 2343, 2366, 2381, See Table 2, I.) (Interferon alfa- cellular responses S, Familletti PC, Pestka S. Chronic Hepatitis;
Hepatitis B; Chronic Hepatitis 2382, 2410, and 3165. SEQ ID NO:Z 0 a,.
2b; recombinant; including antiviral, (1981) Convenient assay for B; Hepatitis C; Chronic Hepatitis C; Hepatitis D; for particular 1 Interferon alfa-nl; antiproliferative, interferons. J. Virol.
Chronic Hepatitis D; Human Papillomavirus;
construct. c7, Interferon alfa-n3; antitumor and 37(2):755-8; Anti-Herpes Simplex Virus Infection;
External "
H
Peginterferon immunomodulatory proliferation assay: Gao Y, Condylomata Acuminata; HIV; HIV Infection;
alpha-2b; activities; stimulate et al (1999) Sensitivity of Oncology;
Cancer; Solid Tumors; Melanoma;
Ribavirin and production of two an epstein-barr virus- Malignant Melanoma; Renal Cancer (e.g., Renal interferon alfa-2b; enzymes: a protein lcinase positive tumor line, Daudi, Cell Carcinoma); Lung Cancer (e.g,. Non-Small Interferon and an oligoadenylate to alpha interferon correlates Cell Lung Cancer or Small Cell Lung Cancer) alfacon-1; synthetase. with expression of a GC- Colon Cancer;
Breast Cancer; Liver Cancer;
interferon rich viral transcript. Mol Prostate Cancer; Bladder Cancer; Gastric Cancer;
consensus; YM Cell Biol. 19(11):7305-13. Sarcoma; AIDS-Related Kaposi's Sarcoma; Iv n 643; CIFN; Lymphoma; T Cell Lymphoma; Cutaneous T- 1-3 interferon -alpha Cell Lymphoma; Non-Hodgkin's Lymphoma;
cp consensus; Brain Cancer; Glioma;
Glioblastoma Multiforrne; o recombinant Cervical Dysplasia;
Leukemia; Preleukemia;

o methionyl Bone Marrow Disorders;
Bone Disorders; Hairy oe o 1--, consensus Cell Leukemia; Chronic Myelogeonus Leukemia;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z -a-, =
interferon; Hematological Malignancies; Hematological o recombinant Disorders; Multiple Myeloma; Bacterial 1--, consensus Infections;
Chemoprotection; Thrombocytopenia;
interferon; CGP Multiple Sclerosis;
Pulmonary Fibrosis; Age-35269; RO Related Macular Degeneration; Macular 253036; RU Degeneration; Crohn's Disease; Neurological 258310; 1NTRON Disorders; Arthritis;
Rheumatoid Arthritis;
A; PEG- Ulcerative Colitis;
Osteoporosis, Osteopenia, INTRON; 01F; Osteoclastogenesis;
Fibromyalgia; Sjogren's =
n OMNIFERON; Syndrome; Chronic Fatigue Syndrome; Fever;
PEG- Hemmorhagic Fever; Viral Hemmorhagic Fevers; 2 OMN1FERON; Hyperglycemia; Diabetes;
Diabetes Insipidus; a, -.3 VELDONA; Diabetes mellitus; Type 1 diabetes; Type 2 H
u.) PEG- diabetes; Insulin resistance; Insulin deficiency; c7, co u at ' = ) REBETRON; Hyperlipidemia;
Hyperketonemia; Non-insulin 10) ROFERON A; dependent Diabetes Mellitus (NIDDM); Insulin- 0 a, WELLFERON; dependent Diabetes Mellitus (IDDM); A 1 ALFERON Condition Associated With Diabetes Including, c7, I.) N/LDO; But Not Limited To Obesity, Heart Disease, H
REBETRON; Hyperglycemia, Infections, Retinopathy, And/Or ALTEMOL; Ulcers; Metabolic Disorders; Immune Disorders;
VIRAFERONPE Obesity; Vascular Disorders; Suppression of G; PEGASYS; Body Weight; Suppression of Appetite;
VIRAFERON; Syndrome X.
VIRAFON;
AMPLIGEN;
Iv INFERGEN;
n INFAREX;

ORAGEN) cp o Calcitonin Regulates levels of Hypocalcemic Rat Bioassay, Bone Disorders; Fracture prevention;
1833, 1834, 1835, 1836, See Table 2, t-.) 1--, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Calcitonin effect opposite to that of stimulation assay: J Bone osteonecrosis; periodontal bone loss;
human-salmon human parathyroid Miner Res 1999 osteoarthritis; rheumatoid arthritis; osteopetrosis;
hybrid; hormone. Aug;14(8):1425-31 periodontal, 13/tic, or metastatic bone disease;
Forcaltonin; osteoclast differentiation inhibition; bone Fortical; disorders; bone healing and regeneration.
Calcitonin;Calcito nina Almirall;
Calcitonina Hubber;
Calcimar;Calsyna r; Calogen; ' Miacalcic;
Miacalcin;
SB205614;
c7, Macritonin;

Cibacalcin;

Cibacalcina;

Cibacalcine;
c7, Salmocalcin;
PowderJect Calcitonin) (CAS-21215-62-3) Interferon beta Modulates MHC antigen Anti-viral assay: Rubinstein Multiple Sclerosis;
Oncology; Cancer; Solid 1778, 1779, 2011, 2013, See Table 2, (Interferon beta- expression, NK cell S, Familletti PC, Pestka S. Tumors; Melanoma;
Malignant Melanoma; Renal 2053, 2054, 2492, 2580, SEQ ID NO:Z
la; Interferon activity and IFNg (1981) Convenient assay for Cancer (e.g., Renal Cell Carcinoma); Lung 2795, 2796, 2797. for particular beta lb; production and IL12 interferons. J. Virol.
Cancer (e.g,. Non-Small Cell Lung Cancer or construct.

Interferon-beta- production in monocytes. 37(2):755-8; Anti- Small Cell Lung Cancer) Colon Cancer; Breast serine; SH 579; proliferation assay: Gao Y, Cancer; Liver Cancer; Prostate Cancer; Bladder ZK 157046; et al (1999) Sensitivity of Cancer; Gastric Cancer; Sarcoma; AIDS-Related BCDF; beta-2 IF; an epstein-barr virus- Kaposi's Sarcoma;
Lymphoma; T Cell oe Interferon-beta-2; positive tumor line, Daudi, Lymphoma;
Cutaneous T-Cell Lymphoma; Non-rhIL-6; SJ0031; to alpha interferon correlates Hodgkin's Lymphoma; Brain Cancer; Glioma;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
DL 8234; with expression of a GC- Glioblastoma Multiforme; Cervical Dysplasia;
PERON; IFNbeta; rich viral transcript. Mol Leukemia;
Preleukemia; Bone Marrow BETASERON; Cell Biol. 19(11):7305-13. Disorders; Bone Disorders; Hairy Cell Leukemia;
AVONEX; Chronic Myelogeonus Leukemia; Hematological REBIF; Malignancies;
Hematological Disorders; Multiple BETAFERON; Myeloma; Bacterial Infections;
SIGOSIX) Chemoprotection;
Thrombocytopenia; Viral infections; HIV Infections; Hepatitis; Chronic Hepatitis; Hepatitis B; Chronic Hepatitis B;
Hepatitis C; Chronic Hepatitis C; Hepatitis D;
Chronic Hepatitis D; Human Papillomavirus;

Herpes Simplex Virus Infection; External Condylomata Acuminata; HIV; HIV Infection;
Pulmonary Fibrosis; Age-Related Macular c7, Degeneration; Macular Degeneration; Crohn's Disease; Neurological Disorders; Arthritis;

Rheumatoid Arthritis; Ulcerative Colitis;

Osteoporosis, Osteopenia, Osteoclastogenesis;
c7, Fibromyalgia; Sjogren's Syndrome; Chronic Fatigue Syndrome; Fever; Hemmorhagic Fever;
Viral Hemmorhagic Fevers; Hyperglycemia;
Diabetes; Diabetes Insipidus; Diabetes mellitus;
Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia;
Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
oe Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;

Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z
o =
Syndrome X.
o Growth hormone Acts on the anterior Growth hormone-releasing Acromegaly; Growth failure; Growth hormone 1747 and 1748. See Table 2, 1--, releasing factor; pituitary to stimulate the peptides (GHRPs) are replacement; Growth hormone deficiency; SEQ ID
NO:Z
Growth hormone production and secretion known to release growth Pediatric Growth Hormone Deficiency; Adult for particular releasing of growth hormone and hormone (GH) in vivo and Growth Hormone Deficiency; Idiopathic Growth construct.
hormone exert a trophic effect on in vitro by a direct action on Hormone Deficiency; Growth retardation;
(Sermorelin the gland. receptors in anterior Prader-Willi Syndrome; Prader-Willi Syndrome acetate; pituitary cells. Biological in children 2 years or older; Growth deficiencies;
Pralmorelin; activity can be measured in Growth failure associated with chronic renal n Somatorelin; cell lines expressing growth insufficiency;
Osteoporosis; Osteopenia, Somatoliberin; hormone releasing factor Osteoclastogenesis;
Postmenopausal 0 I.) Geref; Gerel; receptor (Mol Endocrinol osteoporosis; bums; Cachexia;
Cancer Cachexia; a, -.3 Groliberin) 1992 Oct;6(10):1734-44, Dwarfism; Metabolic Disorders;
Obesity; Renal H
u.) c7, Molecular Endocrinology, failure; Turner's Syndrome; Fibromyalgia;
u.) t.) Vol 7, 77-84). Fracture treatment;
Frailty, AIDS wasting; I.) c:3 Muscle Wasting; Short Stature; Diagnostic a, ' Agents; Female Infertility; lipodystrophy.

IL-2 Promotes the growth of B T cell proliferation assay Cancer; Solid Tumors; Metastatic Renal Cell 1757, 1758, 1812, 1813, See Table 2, c7, I.) (Aldesleukin; and T cells and augments "Biological activity of Carcinoma; Metastatic Melanoma; Malignant 1952, 1954, 2030, and 2031. SEQ ID NO:Z H
cell growth factor; Escherichia coli." Science Lung Cancer or Small Cell Lung Cancer); Colon PROLEUKIN; 223: 1412-1415, 1984. Cancer; Breast Cancer; Liver Cancer; Leukemia;
IMMUNACE; natural killer (NK) cell and Preleukemia;
Hematological Malignancies;
CELEUK; CTL cytotoxicity assay Hematological Disorders; Acute Myeloid ONCOLLP1N 2; "Control of homeostasis of Leukemia;
Melanoma; Malignant Melanoma;
Iv MACROLIN) CD8+ memory T cells by Non-Hodgkin's Lymphoma; Ovarian Cancer; n opposing cytokines. Science Prostate Cancer; Brain Cancer; Glioma;
288: 675-678, 2000; CTLL- Glioblastoma Multiforme; Hepatitis; Hepatitis C;
cp o 2 Proliferation: Gillis et al Lymphoma; HIV Infection (AIDS);
t-.) (1978) J. J. Immunol. 120, Inflammatory Bowel Disorders; Kaposi's o oe 2027 Sarcoma; Multiple Sclerosis; Arthritis; o 1--, Rheumatoid Arthritis; Transplant Rejection;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein=Z -a-, =
=
Diabetes; Type 1 Diabetes Mellitus; Type 2 =

Diabetes.
1--, Parathyroid Acts in conjuction with Adenylyl cyclase Bone Disorders; Fracture prevention; 1749, 1750, 1853, 1854, See Table 2, hormone; calcitonin to control stimulation in rat Hypercalcemia; Malignant hypercalcemia; 1889, 1906, 1907, 1914, SEQ ID NO:Z
parathyrin (PTH; calcium and phosphate osteosarcoma cells, Osteoporosis; Paget's disease; Osteopenia, 1932, 1938, 1941, 1949, for particular Ostabolin; ALX1- metabolism; elevates ovariectomized rat model of Osteoclastogenesis; osteolysis;
osteomyelitis; 2021, 2022, 2023, 2428, construct.
11; hPTH 1-34; blood calcium level;
osteoporosis: IUBMB Life osteonecrosis; periodontal bone loss;
2714, 2791, 2965, 2966.
LY 333334; MN stimulates the activity of 2000 Feb;49(2):131-5.
osteoarthritis; rheumatoid arthritis; osteopetrosis;
10T; parathyroid osteocytes; enhances periodontal, lytic, or metastatic bone disease;
n hormone (1-31); absorption of Ca+/Pi from osteoclast differentiation inhibition; bone FORTEO; small intestine into blood; disorders; bone healing and regeneration. 0 I.) PARATHAR) promotes reabsorption of a, -.3 Ca+ and inhibits Pi by H
CA
c7, kidney tubules.
u.) t .) .,...1 Resistin Mediates insulin Ability of resistin to Hyperglycemia; Diabetes; Diabetes Insipidus; 2295, 2296, 2297, 2300, See Table 2, "

resistance in Type II influence type II diabetes Diabetes mellitus; Type 1 diabetes; Type 2 and 2309. SEQ ID NO:Z

a, I
diabetes; inhibits insulin- can be determined using diabetes; Insulin resistance; Insulin deficiency; for particular 0 c7, I.) uptake Pontoglio et al., J Chin dependent Diabetes Mellitus (NIDDM); Insulin- H
Invest 1998 May dependent Diabetes Mellitus (IDDM); A
15;101(10):2215-22. Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Iv Syndrome X.
n o 3; FASTR) mediated apoptosis. staining, TUNEL
staining, fulminant liver failure); graft versus host 1645, 1700, 1702, 1703, for particular t-.) levels. Inhibition of cell syndrome; renal failure;
insulin dependent and 1913. vD
1-, growth can also be directly diabetes mellitus;
rheumatoid arthritis;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z -a-, =
measured, for example by inflammatory bowel disease; autoimmune =
--õi ALOMAR Blue staining, disease; toxic epidermal necrolysis; multiple 1--, Assay refs: cytotoxicity sclerosis.
assay on human fibrosarcoma (Epsevik and Nissen-Meyer, 1986, J.
Immunol. methods).
, DeCAF (D- Inhibits proliferation DeCAF activity can be B cell and/or T cell mediated immune 1657. See Table 2, SLAM; BCM- and differentiation of B determined using assays disorders; Immunodeficiency (e.g., Common SEQ
ID NO:Z
n like membrane cells; Antagonize BLyS known in the art, such as for Variable Immunodeficiency, Selective IgA for particular protein; activity example, those described in Deficiency) construct. 2 BLAME (B Examples 32-33 of ti H
lymphocyte International Publication u.) activator No. W00111046.
c7, u.) I.) macrophage IO) oo .
expressed)) BLyS (B Promotes proliferation, BLyS activity can be B cell and/or T cell mediated immune 1680, 2095, and 2096. See Table 2, 1 co c7, Lymphocyte differentiation and determined using assays disorders, particularly immune system SEQ ID
NO:Z 1 I.) Stimulator; survival of B cells; known in the art, such as, disorders associated with low B cell numbers for particular H
Neutrokine Promotes for example, the or low serum immunoglobulin; construct.
alpha; TL7; immunoglobulin costimulatory proliferation Immunodeficiency (e.g., Common Variable BAFF; TALL- production by B cells, assay and other assays Inimunodeficiency, Selective IgA Deficiency).
1; THANK; disclosed by Moore et al., Radiolabeled forms: lymphoma, non-Hodgkins radiolabeled 1999, Science, lymphoma, chronic lymphocytic leukemia, BLyS) 285(5425):260-3. multiple myeloma.
Anti-BLyS Agonize or antagonize BLyS agonist or antagonist B cell and/or T cell mediated immune 1821, 1956, 2501, 2502, See Table 2, Iv single chain BlyS activity, activity can be determined disorders; Autoimmune disorders, particularly 2638. SEQ ID NO:Z
n antibody (sc using assays known in the autoimmune diseases associated with the for particular EvIl 16A01, art, such as, for example, a production of autoantibodies; Rheumatoid construct. cp scEvI050B11, modified version the Arthritis, Systemic Lupus Erythmatosus;
scEvI006D08) costimulatory proliferation Sjogren's Syndrome, cancers expressing Blys a and others, assay disclosed by Moore et as an autocrine growth factor, e.g. certain vD
1--, al., 1999, Science, chronic lymphocytic leukemias.
, =
Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a, Protein:X Assay Protein:Z
=
285(5425):260-3, in which =

BlyS is mixed or 1--, preincubated with the anti-BlyS antibody prior to being applied to the responder B lymphocytes.
MPIF-1 Inhibits myeloid MPIF-1 activity can be Chemoprotection; Adjunct to Chemotherapy; 1681, 3166, 3167, 3168, See Table 2, (Myeloid progenitor cells; and measured using the Inflammatory disorders; Cancer; Leukemia; SEQ
ID NO:Z
Progenitor activates monocytes myeloprotection assay and Myelocytic leukemia; Neutropenia, Primary for particular n Inhibitory chemotaxis assay described neutropenias (e.g.; Kostmann syndrome); construct.
Factor; CK in US patent 6,001,606.
Secondary neutropenia;
Prevention of 0 I.) beta-8; neutropenia; Prevention and treatment of a, -.3 Mirostipen) neutropenia in HIV-infected patients; H
CA
c7, Prevention and treatment of neutropenia u.) t\.) associated with chemotherapy; Infections N) vD

associated with neutropenias; Myelopysplasia;

a, Autoimmune disorders; Psoriasis; Mobilization c7, of hematopoietic progenitor cells; Wound I.) Healing; Autoimmune Disease; Transplants;
H
Bone marrow transplants; Acute myelogeneous leukemia; Lymphoma, Non-Hodgkin's lymphoma; Acute lymphoblastic leukemia;
Hodgkin's disease; Accelerated myeloid recovery; Glycogen storage disease.
KDI Inhibits bone marrow KDI activity can be Multiple sclerosis; Hepatitis; Cancer; Viral 1746. See Table 2, (Keratinocyte proliferation; and shows measured using the antiviral infections, HIV infections, Leukemia. SEQ ID NO:Z Iv Derived antiviral activity, and cell proliferation assays for particular n 1-i Interferon; described in Examples 57-construct.
Interferon 63 of International cp o Kappa Publication No.
t-.) Precursor) WOO W00107608.
a - TNFR2 (p75) Binds both TNFa and T-cell proliferation can be Autoimmune disease; Rheumatoid Arthritis; 1777 and 1784. See Table 2, vD
1--, (ENBREL) TNFb; mediates T-cell measured using assays Psoriatic arthritis; Still's Disease; Ankylosing SEQ ID NO:Z

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z -a-, =
proliferation by TNF; known in the art. For Spondylitis; Cardiovascular Diseases; for particular o reduces signs and example, "Lymphocytes: a Vasulitis; Wegener's granulomatosis; construct.
1--, structural damage in practical approach" edited Amyloidosis;
Systemic Lupus Erythematosus, patients with by: SL Rowland, AJ Insulin-Dependent Diabetes Mellitus;
moderately to severly McMichael ¨ chapter 6, Immunodeficiency Disorders; Infection;
active rheumatoid pages 138-160 Oxford Inflammation;
Inflammatory Bowel Disease;
arthritis (RA). University Press (2000); Chrohn's Disease;
Psoriasis; AIDS; Graft and "Current Protocols on Rejection; Graft Versus Host Disease.
CD-ROM" section 3.12 n Proliferation Assays for T-cell Function John Wiley I.) & Soncs, Inc. (1999).
a, -.3 Keratinocyte Stimulates epithelial KGF-2 activity can be Stimulate Epithelial Cell Proliferation; 1785, 1786, 1916, 1917, See Table 2, H
CA
.
c7, growth factor 2 cell growth. measured using the wound Stimulate Basal Keratinocytes; Wound 2498, 2499, 2552, 2553, SEQ
ID NO:Z u.) (Repifermin; healing assays and Healing; Stimulate Hair Follicle Production; 2584, 2607, 2608, 2606, for particular N) KGF-2; epithelial cell proliferation Healing Of Dermal Wounds. Wound Healing; 2630 construct. 0 a, Fibroblast assays described in US
Eye Tissue Wounds, Dental Tissue Wounds, 1 c7, Growth Factor- patent 6,077,692. Oral Cavity Wounds, Diabetic Ulcers, Dermal 1 I.) 10; FGF-10) Ulcers, Cubitus Ulcers, Arterial Ulcers, Venous H
Stasis Ulcers, Burns Resulting From Heat Exposure Or Chemicals, or Other Abnormal Wound Healing Conditions such as Uremia, Malnutrition, Vitamin Deficiencies or Complications Associated With Systemic Treatment With Steroids, Radiation Therapy or Antineoplastic Drugs or Antimetabolites;
Iv Promote Dermal Reestablishment Subsequent n To Dermal Loss; Increase the Adherence Of Skin Grafts To A Wound Bed; Stimulate Re-cp o Epithelialization from The Wound Bed; To t-.) Promote Skin Skin Strength; Improve The a Appearance Of Aged Skin; Proliferate vD
1--, Hepatocytes, Lung, Breast, Pancreas, Stomach, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Bladder, Small Intestine, Large Intestine;
Sebocytes, Hair Follicles, Type II
Pneumocytes, Mucin-Producing Goblet Cells, or Other Epithelial Cells, Endothelial Cells, Keratinocytes, or Basal Keratinocytes (and Their Progenitors) Contained Within The Skin, Lung, Liver, Bladder, Eye, Salivary Glands, or Gastrointestinal Tract; Reduce The Side Effects Of Gut Toxicity That Result From Radiation, Chemotherapy Treatments Or Viral Infections; Cytoprotector, especially of the Small Intestine Mucosa or Bladder; Mucositis (Mouth Ulcers); Regeneration Of Skin; Full c7, and/or Partial Thickness Skin Defects, including Bums, (e.g., Repopulation Of Hair Follicles, Sweat Glands, And Sebaceous Glands); Psoriasis; Epidermolysis Bullosa;

c7, Blisters; Gastric and/or Doudenal Ulcers;
Reduce Scarring; Inflamamatory Bowel Diseases; Crohn's Disease; Ulcerative Colitis;
Gut Toxicity; Lung Damage; Repair Of Alveoli And/or Brochiolar Epithelium; Acute Or Chronic Lung Damage; Emphysema, ARDS;
Inhalation Injuries; Hyaline Membrane Diseases; Infant Respiratory Distress Syndrome; Bronchopulmonary Displasia In Premature Infants; Fulminant Liver Failure;
Cirrhosis, Liver Damage caused by Viral Hepatitis and/or Toxic Substances; Diabetes Mellitus; Inflammation.
TR2svl, proliferation, and proliferation assay and Ig disease; graft versus host disease; graft SEQ ID
NO:Z
TR2SV2; mediates and inhibits production assay (Moore et rejection; variable immunodeficiency; for particular Table 1 _______________________________________________________________________________ ________________________________________ 0 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z 'a =
TNFRSF14; Herpes Simplex Virus al., 1999, Science, immunodeficiency syndromes; cancer. construct.
o HVEM; Herpes (HSV) infection. 285(5425):260-3.). HSV-1 1--, Virus Entry and HSV-2 Infectivity Mediator; Assay: International ATAR) Publication No. WO

Macrophage Chemotactic for Chemokine activities can be Inflammatory diseases; wound healing; 1809, 2137, 2474, 2475, See Table 2, derived monocyte-derived determined using assays angiogenesis; AIDS infection. 2476, and 2477. SEQ ID NO:Z
chemolcine, dendritic cells and IL-2- known in the art: Methods for particular IVIDC (Ckbeta- activated natural killer in Molecular Biology, construct.
13) cells. 2000, vol. 138: Chemokine I.) Protocols. Edited by: A.E.I.
a, -.3 Proudfoot, T.N.C. Wells, H
CA
c7, and C.A. Power. 0 Humana u.) (k) Press Inc., Totowa, NJ
I.) t.) a, ' HAGDG59 Activates MIPla Dendritic cell assays are Immune disorders; cancer; viral infection; 1830 and 1831. See Table 2, 0 (Retinal short- release in Dendritic well known in the art.
For inflammation; sepsis; arthritis; asthma. SEQ
ID NO:Z c7, I.) chain Cells. example, J. Immunol.
for particular H
dehydrogenase) 158:2919-2925 (1997); J.
construct.
Leukoc. Biol. 65:822-828 (1999).
GnRH Promotes release of GnRH is known to cause Infertility; Kallmann's syndrome or other forms 1862 and 1863. See Table 2, (Gonadotropin follicle-stimulating the release of follicle of hypergonadotropic hypergonadism (failure SEQ
ID NO:Z
Releasing hormone and luteini7ing stimulating hormone (FSH) to go through puberty naturally). for particular Hormone) hormone from anterior and/or luteinizing hormone construct.
Iv pituitary. (LH) in vivo by a direct n ,-i action on receptors in anterior pituitary cp o gonadotropes. GnRH
t-.) activity can can be determined o co) by measuring FSH levels in o 1--, the medium of cultured Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z 'a o o gonadotropes before and o after GnRH
1--, supplementation. For example, Baker et al. Biol Reprod 2000 Sep;63(3):865-71.
Teprotide Inhibits angiotensin Inhibition of ACE can be Hypertension; congestive heart failure. 1866, 1867, 2025, and See Table 2, converting enzyme determined using assays 2026. SEQ ID NO:Z
(ACE). known in the art. For for particular n example, Anzenbacherova construct.
et al., J.Phaima Biomed I.) Anal 2001 Mar; 24(5-a, -.3 6):1151-6.
H
u.) Human Involved in Chemokine activities can be Autoimmune disorders; Immunity; Vascular 1933, 1934, 1947, 1948, See Table 2, u.) Lk.) u.) chemokine inflammation, allergy, determined using assays and Inflammatory disorders; HIV; AIDS; 1955, 1998, 2355, 2412, SEQ ID NO:Z I.) HCC-1 (ckBeta- tissue rejection, viral known in the art:
Methods infectious diseases. 2449, 2837, 2838, 2839, for particular 0 a, 1; HWFBD) infection, and tumor in Molecular Biology, 2840, 2841, 2842, 2843, construct. 0 biology; enhances 2000, vol. 138: Chemokine 2844, 2845, 2849, 2947, , proliferation of CD34+ Protocols. Edited by: A.E.I.
3066, 3105, 3124, 3125, I.) H
myeloid progenitor Proudfoot, T.N.C. Wells, 3139, 3152, 3153, 3154, cells. and C.A. Power. Humana 3155, 3156, 3169, 3170, Press Inc., Totowa, NJ
3202, 3203, 3204, 3205, 3206,3207,3272 ACE2 inhibitor Inhibits production of Inhibition of angiotensin Treatment for elevated angiotensin II and/or 1989, 2000, 2001, and See Table 2, (DX512) angiotensin II which can be determined using aldosterone levels, which can lead to 2002. SEQ ID NO:Z
induces aldosterone assays known in the art.
vasoconstriction, impaired cardiac output for particular Iv production, arteriolar For example, in vitro using and/or hypertension; Cardiovascular Disease;
construct. n smooth muscle a proliferation assay with Cardiac Failure; Diabetes; Type II
Diabetes; 1-3 vasoconstriction, and rat cardiac fibroblasts as Proteinuria; Renal disorders, congestive heart cp o proliferation of cardiac described in Naunyn failure.
t-.) fibroblasts, Induces Induces Schmiedebergs Arch o oe angiogenesis; an Pharmacol 1999 o 1--, enzyme that converts May;359(5):394-9.

, Table 1 -o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay Protein:Z -a-, =
angiotensin Ito o angiotensin1-9; also cleaves des-Arg, bradykinin and neurotensin.
TR1 (OCIF; Inhibits Coculture Assay for Osteoporosis; Paget's disease; osteopenia; 2016, 2017, 2085, 2086, See Table 2, Osteoclastogene osteoclastogenesis and Osteoclastogenesis, Bone osteolysis; osteomyelitis; osteonecrosis; 2529, 2530, 2531, 2532, SEQ ID NO:Z
sis inhibitory bone resorption, and resorption assay using fetal periodontal bone loss; osteoarthritis; 2555, 2556, 2557, and for particular factor; induces fibroblast long-bone organ culture rheumatoid arthritis; osteopetrosis; periodontal, 2558. construct.
n osteoprotegerin, proliferation, system, dentine resorption lytic, or metastatic bone disease; osteoclast OPG; tumor assay, and fibroblast differentiation inhibition;
bone disorders; bone 0 I.) necrosis factor proliferation-assays are healing and regeneration; organ calcification; a, -.3 receptor each described in Kwon et vascular calcification. H
CA
superfamily al., FASEB J. 12: 845-854 c7, u.) member 11B (1998).
I.) 4=, precursor;) a, Human Chemotactic for both Chemokine activities can be Cancer; Wound healing; Inflammatory 2101, 2240, 2241, 2245, See Table 2, 1 chemokine activated (CD3+) T determined using assays disorders; Immmunoregulatory disorders; 2246, 2247, and 2248.
SEQ ID NO:Z c7, I.) (CD14-) lymphocytes in Molecular Biology, Parasitic Infection; Rheumatoid Arthritis;
construct.
and (CD4+) and 2000, vol. 138: Chemokine Asthma; Autoimmune disorders.
(CD 8+) T lymphocytes Protocols. Edited by: A.E.I.
and (CD45RA+) T cells Proudfoot, T.N.C. Wells, and C.A. Power. Humana Press Inc., Totowa, NJ
Iv (HGBAN46; microbicidal determined using assays Autoimmune Disorders; Psoriasis; Asthma; 2337, 2338, and 2348.
SEQ ID NO:Z n HE9DR66) leukocytes; Attracts known in the art:
Methods Allergy; Hematopoiesis; Wound Healing; Bone for particular 1-3 o immature dendritic cells 2000, vol. 138: Chemokine Eosinophilic Syndrome; Lung Inflammation; t-.) and memory/effector T Protocols. Edited by: A.E.I. Fibrotic Disorders;
Atherosclerosis; Periodontal a cells; B-cell Proudfoot, T.N.C. Wells, diseases; Viral diseases; Hepatitis.
o 1-, chemotaxis; inhibits and C.A. Power. Humana Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay -Protein:Z -a-, proliferation of myeloid Press Inc., Totowa, NJ
o o progenitors; chemotaxis 1--, of PBMC's. .
Leptin Controls obesity in vivo modulation of food Hyperglycemia; Diabetes; Diabetes Insipidus; 2146, 2184, 2186, and See Table 2, through regulation of intake, reduction in body Diabetes mellitus; Type 1 diabetes; Type 2 2187. SEQ ID NO:Z
appetite, reduction of weight, and lowering of diabetes; Insulin resistance; Insulin deficiency;
for particular body weight, and insulin and glucose levels in Hyperlipidemia; Hyperketonemia; Non-insulin construct.
lowering of insulin and ob/ob mice, dependent Diabetes Mellitus (NIDDM);
and activation of the leptin Condition Associated With Diabetes Including, n receptor in a cell-based But Not Limited To Obesity, Heart Disease, 0 I.) assay. Protein Expr Purif Hyperglycemia, Infections, Retinopathy, a, -.3 1998 Dec;14(3):335-42 And/Or Ulcers; Metabolic Disorders; Immune H
CA
Disorders; Obesity; Vascular Disorders;
c7, u.) Lk.) LA Suppression of Body Weight; Suppression of I.) Appetite; Syndrome X; Immunological a, Disorders; Immunosuppression.

IL-1 receptor Binds IL1 receptor 1) Competition for IL-1 Autoimmune Disease; Arthritis; Rheumatoid 2181, 2182, 2183, and See Table 2, c7, antagonist without activating the oinding to IL-1 receptors in Arthritis; Asthma;
Diabetes; Diabetes Mellitus; 2185. SEQ ID NO:Z I.) H
(Anakirffa; target cells; inhibits the YT-NCI or C3H/HeJ cells GVHD; Inflammatory Bowel Disorders; for particular interleukin-1 and ILl-beta; and 633-638, 1990); Psoriasis; Septic Shock; Transplant Rejection;
receptor; TRAP; neutralizes the biologic Inflammatory Disorders;
Rheumatic Disorders;
KINERET; activity of IL1-alpha 2) Inhibition of IL-1-Osteoporosis; Postmenopausal Osteoporosis;
ANTRIL) and IL1-beta. induced endothelial cell- Stroke.
Leukocyte adhesion (Carter 3t al., Nature 344: 633-638, od n 1990);

cp o 3) Proliferation assays on A375-C6 cells, a human o oe melanoma cell line highly o 1--, susceptible to the Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z -a-, antiproliferative action of =
o IL-1 (Murai T et al., J. Biol.

1-, Chem. 276: 6797-6806, 2001).
TREM-1 Mediates activation of Secretion of cytokines, Inflammation; Sepsis; bacterial infection; 2226 and 2230. See Table 2, (Triggering neutrophil and chemokines, degranulation, autoimmune diseases; GVHD. SEQ ID NO:Z
Receptor monocytes; Stimulates and cell surface activation for particular Expressed on neutrophil and markers can be determined construct.
Monocytes 1) monocyte-mediated using assays described in inflammatory response; Bouchon et al, J Immunol n Promotes secretion of 2000 May TNF, IL-8, and MCP-1; 15;164(10):4991-5.
"
a, -.3 Induces neutrophil H
u.) degranulation, Ca2+
c7, u.) La mobilization and c:3 iv tyrosine phosphorylation of a, extracellular signal-c7, I
related kinase 1 iv H
(ERK1), ERK2 and phospholipase C-gamma.
HCNCA73 Induces T-cell FMAT can be used to Autoimmune disorders;
Inflammation of the 2244 and 2365. See Table 2, activation- expression measure T-cell surface gastrointestinal tract;
Cancer; Colon Cancer; SEQ ID NO:Z
of CD152 marker; markers (CD69, CD152, Allergy; Crohn's disease, for particular Stimulates release of CD71, HLA-DR) and T-cell construct.
TNF-a and MIP-la cytokine production (e.g., Iv n from immature, IFNg production). J. of monocyte-derived Biomol. Screen. 4:193-204 cp dendritic cells; (1999). Other T-cell Promotes maturation of proliferation assays:

o dendritic cells. "Lymphocytes: a practical oe o 1-, approach" edited by: SL

Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay Protein:Z -a-, Rowland, AJ McMichael -=
o --I
Chapter 6, pages 138-160 Oxford University Press (2000); WO 01/21658 Examples 11-14, 16-17 and 33.
VEGF-2 Promotes endothelial VEGF activity can be Coronary artery disease; Critical limb ischemia;
2251, 2252, 2256, and See Table 2, (Vascular cell proliferation, determined using assays Vascular disease; proliferation of endothelial 2257. SEQ ID NO:Z
Endothelial known in the art, such as cells, both vascular and lymphatic. Antagonists for particular Growth Factor- those disclosed in may be useful as anti-angiogenic agents; construct. n 2; VEGF-C) International Publication Cancer. 0 I.) No. W00045835, for a, -.3 example.
H
CA
HCHNF25 Activates MIPla Dendritic cell assays are Immune disorders; cancer. 2271, 2280, and 2320. See Table 2, c7, u.) (jumping Release in Dendritic well known in the art. For SEQ ID NO:Z I.) translocation Cells. example, J. Immunol.
for particular 0 a, breakpoint) 158:2919-2925 (1997); J.
construct. 0 Leukoc. Biol. 65:822-828 c7, I.) (1999).
H
HLDOU18 Activates L6/GSK3 Assays for activation of Hyperglycemia; Diabetes; Diabetes Insipidus; 2328, 2340, 2350, 2351, ee Table 2, (Bone lcinase assay. GSK3 kinase activity are Diabetes mellitus;
Type 1 diabetes; Type 2 2359, 2362, 2367, 2369, EQ ID NO:Z
Morphogenic well known in the art. For diabetes; Insulin resistance; Insulin deficiency; 2370, 2473, or particular Protein 9 example, Biol. Chem.
Hyperlipidemia; Hyperketonemia; Non-insulin 2623, 2624, onstruct.
(BMP9); 379(8-9): (1998) 1101- dependent Diabetes Mellitus (NIDDM); Insulin- 2625, 2631, Growth 1110.; Biochem J. 1993 dependent Diabetes Mellitus (IDDM); A 2632, 2633.
differentiation Nov 15;296 ( Pt 1):15-9. Condition Associated With Diabetes Including, factor-2 But Not Limited To Obesity, Heart Disease, n precursor Hyperglycemia, Infections, Retinopathy, 1-3 (GDF-2 And/Or Ulcers; Metabolic Disorders; Immune cp o precursor)) Disorders; Obesity;
Vascular Disorders; t-.) Suppression of of Body Weight; Suppression of o oe Appetite; Syndrome X.
vD
1-, Glucagon-Like- Stimulates the synthesis GLP1 activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; 2448, 2455, 2456, 2457, See Table 2, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z 'a Peptide 1 (GLP1; and release of insulin;
assayed in vitro using a [3- Diabetes mellitus; Type 1 diabetes; Type 2 2803, 2804, 2900, 2904, SEQ ID NO:Z
=

Insulinotropin) enhances the sensitivity of H]-glucose uptake assay. (J
diabetes; Insulin resistance; Insulin deficiency; 2945, 2964, 2982, 3070, for particular 1--, adipose, muscle, and liver Biol Chem 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non-insulin 2802, 3027, 3028, 3045, construct.
tissues towards insulin; 274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin- 3046, 3069, 3071, 3072, stimulates glucose uptake; dependent Diabetes Mellitus (IDDM); A 3085, 3086, 3087, 3140, slows the digestive Condition Associated With Diabetes Including, 3309 process; suppresses But Not Limited To Obesity, Heart Disease, appetite; blocks the Hyperglycemia, Infections, Retinopathy, And/Or secretion of glucagon. Ulcers; Metabolic Disorders; Immune Disorders;
n Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;

I.) Syndrome X.
a, -.3 H
Exendin-4 (AC- Stimulates the synthesis Exendin-4 activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; 2469 and 2470.
See Table 2, u.) c7, 2993) and release of insulin;
assayed in vitro using a [3- Diabetes mellitus;
Type 1 diabetes; Type 2 SEQ ID NO:Z u.) kJ.>
"
oo enhances the sensitivity of 1-1]-glucose uptake assay. (J
diabetes; Insulin resistance; Insulin deficiency; for particular 0 adipose, muscle, and liver Biol Chem 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non-insulin construct. a, tissues towards insulin; 274(43):30864-30873).
dependent Diabetes Mellitus (NIDDM); Insulin- 0 c7, stimulates glucose uptake; dependent Diabetes Mellitus (IDDM); A 1 I.) slows the digestive Condition Associated With Diabetes Including, H
process; suppresses But Not Limited To Obesity, Heart Disease, appetite; blocks the Hyperglycemia, Infections, Retinopathy, And/Or secretion of glucagon. Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X.
T20 (T20 HIV a peptide from residues Virus inhibition assays as HIV; AIDS;
SIV (simian immunodeficiency 7777, 2672, 2673 See Table 2, Iv inhibitory 643-678 of the HIV gp41 described in Zhang et al., virus) infection. SEQ ID NO:Z n ,-i peptide, DP178; transmembrane protein Sept. 26 2002, for particular DP178 HIV ectodomain which binds Sciencexpress construct. cp inhibitory to gp41 in its resting state (www.sciencexpress.org).
peptide) and and prevents occ:' vD
transformation to the 1--, fusogenic state Table 1 _______________________________________________________________________________ ________________________________________ o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay Protein:Z
T1249 (T1249 a second generation HIV Virus inhibition assays as HIV; AIDS; SIV (simian immunodeficiency 9999, 2667, 2670, 2946 See Table 2, o HIV inhibitory fusion inbitor described in Zhang et al., virus) infection SEQ ID NO:Z --1 1--, peptide; T1249 Sept. 26 2002, for particular anti-HIV peptide) Sciencexpress construct.
(wwW.sciencexpress.org).
Interferon Confers a range of Anti-viral assay: Rubinstein Viral infections; HIV Infections; Hepatitis; 2875, 2872, 2876, 2874, See Table 2, Hybrids, cellular responses S, Familletti PC, Pestka S. Chronic Hepatitis; Hepatitis B; Chronic Hepatitis 2873. SEQ ID NO:Z
specifically including antiviral, (1981) Convenient assay for B; Hepatitis C; Chronic Hepatitis C; Hepatitis D; for particular preferred: antiproliferative, interferons. J. Virol.
Chronic Hepatitis D; Human Papillomavirus;
construct.
antitumor and 37(2):755-8; Anti- Herpes Simplex Virus Infection; External n IFNalpha AID immunomodulatory proliferation assay: Gao Y, Condylomata Acuminata; HIV; HIV Infection; 0 I.) hybrid (BgIII activities; stimulate et al (1999) Sensitivity of Oncology; Cancer; Solid Tumors; Melanoma;
-.3 version) production of two an epstein-barr virus-Malignant Melanoma; Renal Cancer (e.g., Renal H
CA
IFNalpha AID enzymes: a protein kinase positive tumor line, Daudi, Cell Carcinoma); Lung Cancer (e.g,. Non-Small c7, u.) t) hybrid (PviiII and an oligoadenylate to alpha interferon correlates Cell Lung Cancer or Small Cell Lung Cancer) I.) version) synthetase. Also, with expression of a GC-Colon Cancer; Breast Cancer;
Liver Cancer; 0 a,.

IFNalpha A/F modulates MHC antigen rich viral transcript. Mol Prostate Cancer; Bladder Cancer; Gastric Cancer; 0 hybrid expression, NK cell Cell Biol. 19(11):7305-13.
Sarcoma; AIDS-Related Kaposi's Sarcoma; c7, I.) IFNalpha A/B activity and IFNg Lymphoma; T Cell Lymphoma; Cutaneous T- H
hybrid production and IL12 Cell Lymphoma; Non-Hodgkin's Lymphoma;
IFNbeta 1/alpha production in monocytes. Brain Cancer; Glioma;
Glioblastoma Multiforrne;
D hybrid Cervical Dysplasia;
Leukemia; Preleukemia;
(IFNbeta-1/alpha- Bone Marrow Disorders;
Bone Disorders; Hairy I hybrid)Cell Leukemia; Chronic Myelogeonus Leukemia;
-IFNalpha/beta Hematological Malignancies; Hematological hybrid Disorders; Multiple Myeloma; Bacterial Iv Infections; Chemoprotection; Thrombocytopenia;
n Multiple Sclerosis; Pulmonary Fibrosis; Age-Related Macular Degeneration; Macular cp o Degeneration; Crohn's Disease; Neurological Disorders; Arthritis; Rheumatoid Arthritis;

o oe Ulcerative Colitis; Osteoporosis, Osteopenia, o 1--, Osteoclastogenesis; Fibromyalgia; Sjogren's Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Syndrome; Chronic Fatigue Syndrome; Fever;
Hemmorhagic Fever; Viral Hemmorhagic Fevers;
Hyperglycemia; Diabetes; Diabetes Insipidus;
Diabetes mellitus; Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency;
Hyperlipidemia; Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of c7, Body Weight; Suppression of Appetite;
Syndrome X.

B-type natriuretic stimulates smooth muscle Inhibition of angiotensin can Congestive heart failure; cardiac volume 3119, 8888. See Table 2, peptide (BNP, relaxation and be determined using assays overload;
cardiac decompensation; Cardiac SEQ ID NO:Z

c7, brain natriuretic vasodilation, known in the art, for Failure; Left Ventricular Dysfunction; Dyspnea for particular peptide) natriuresis,and example using an in vitro construct.
suppression of renin- proliferation assay with rat angiotensin and cardiac fibroblasts as endothelin. described in Naunyn Schmiedebergs Arch Pharmacol 1999 May;359(5):394-9.
Vasodilation can be measured in animals by measuring the myogenic responses of small renal arteries in an isobaric arteriograph system (see Am J Physiol Regul Integr Comp Physiol 2002 Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z 'a o Aug;283(2):R349-R355).
=
o Natriuesis is determined by 1--, measuring the amount of sodium in the urine.
cc-defensin, Suppression of HIV Virus inhibition assays as HIV, AIDS; ARC. 3208, 3209, 3210. See Table 2, including alpha 1 replication; active against described in Zhang et al., SEQ ID NO:Z
defensin, alpha 2 bacteria, fungi, and Sept. 26 2002, for particular defensin, alpha 3 enveloped viruses.
Sciencexpress construct.
defensin (www.sciencexpress.org).
(myeloid-related n defensin; DEFAl;

I.) neutrophil-a, -.3 specific defensin;
H
CA
CAF) c7, u.) 4" Phosphatonin Regulation of phosphate Blood phosphate levels can Hyperphosphatemia;
Hyperphosphatemia in 3238. See Table 2, I.) (matrix metabolism, be measured using methods chronic renal failure; hypophosphatemia; SEQ ID NO:Z 0 a, extracellular known in the art such as the Osteomalacia;
Rickets; X-linked dominant for particular 1 phosphoglycoprot Hypophosphatemic Rat hypophosphatemic rickets/osteomalacia (XLH); construct. c7, em; MEPE) Bioassay. Zoolog Sci 1995 autosomal dominant hypophosphatemic "
H
Oct;12(5):607-10. rickets/osteomalacia (ADHR); tumor-induced rickets/osteomalacia (TI).
P lpal-12 Regulation of protease- Platelet aggregation can be Protection against systemic platelet activation, 3274. See Table 2, (pepducin, activated receptor (PAR) measured using methods thrombus, heart attack, stroke, and/or coagulation SEQ ID NO:Z
PAR1-based signal transduction and known in the art such as disorders, for particular pepducin) thrombin-mediated described in Nature construct.
aggregation of human Medicine 2002 Oct; 8(10):
platelets. 1161-1165.
Iv n (pepducin, PAR4- activated receptor (PAR) measured using methods thrombus, heart attack, stroke, and/or coagulation SEQ ID NO:Z
cp o thrombin-mediated described in Nature construct. o oe aggregation of human Medicine 2002 Oct; 8(10):
o 1--, Table 1 c Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay Protein:Z
cA
=
platelets. 1161-1165.
o --.1 HRDFD27 Involved in the T-cell proliferation can be Chemoprotection; Adjunct to Chemotherapy; 2361 See Table 2, 1--, proliferation of T cells; measured using assays Inflammatory disorders;
Cancer; Leukemia; SEQ ID NO:Z
Production of known in the art. For Myelocytic leukemia; Neutropenia, Primary for particular TNFgamma. example, "Lymphocytes: a neutropenias (e.g.;
Kostmann syndrome); construct.
practical approach" edited Secondary neutropenia; Prevention of by: SL Rowland, Al neutropenia; Prevention and treatment of McMichael ¨ chapter 6, neutropenia in HIV-infected patients; Prevention pages 138-160 Oxford and treatment of neutropenia associated with n University Press (2000); and chemotherapy; Infections associated with "Current Protocols on CD- neutropenias; Myelopysplasia; Autoimmune I.) ROM" section 3.12 disorders; Psoriasis;
Mobilization of a, -.3 H
Proliferation Assays for T- hematopoietic progenitor cells; Wound Healing;
u.) c7, cell Function John Wiley & Autoimmune Disease; Transplants; Bone marrow u.) 4=. Soncs, Inc. (1999). transplants; Acute myelogeneous leukemia; N) Iv Lymphoma, Non-Hodgkin's lymphoma; Acute a, lymphoblastic leukemia; Hodgkin's disease;

c7, Accelerated myeloid recovery; Glycogen storage I.) disease H
HWHGZ51 Stimulates an immune The ability to affect Skeletal diseases and disorders; Musculoskeletal 2407, 2408 See Table 2, (CD59; response and induces chondrocyte differentiation diseases and disorders;
Bone fractures and/or SEQ ID NO:Z
Metastasis- inflammation by inducing can be measured using breaks; Osteoporosis (postmenopausal, senile, or for particular associated GPI- mononuclear cell, methods known in the art, idiopathic juvenile); Gout and/or pseudogout;
construct.
adhered protein eosinophil and PMN such as described in Bone Paget's disease; Osteoarthritis; Tumors and/or homolog) infiltration; Inhibits (1995) Sep; 17(3):279-86. cancers of the bone (osteochondromas, benign growth of breast cancer, chondromas, chondroblastomas, chondromyxoid Iv ovarian cancer, leukemia, fibromas, osteoid osteomas, giant cell tumors, n and melanoma; multiple myelomas, osteosarcomas, Overexpressed in colon, fibrosarcomas, malignant fibrous histiocytomas, cp o lung, breast and rectal chondrosarcomas, Ewing's tumors, and/or t-.) tumors; Regulates Regulates glucose malignant lymphomas);
Bone and joint infections a and/or FFA update by (osteomyelitits and/or infectious arthritis);
1--, adipocytes and skeletal Charcot's joints; Heel spurs; Sever's disease;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
muscle; Induces Sport's injuries;
Cancer; Solid Tumors;
redifferentiation of Melanoma; Malignant Melanoma; Renal Cancer chondrocytes (e.g., Renal Cell Carcinoma); Lung Cancer (e.g,.
Non-Small Cell Lung Cancer or Small Cell Lung Cancer) Colon Cancer; Breast Cancer; Liver Cancer; Prostate Cancer; Bladder Cancer; Gastric Cancer; Sarcoma; AIDS-Related Kaposi's Sarcoma; Lymphoma; T Cell Lymphoma;
Cutaneous T-Cell Lymphoma; Non-Hodgkin's Lymphoma; Brain Cancer; Glioma; Glioblastoma Multiforme; Cervical Dysplasia; Leukemia;

Preleukemia; Bone Marrow Disorders; Bone Disorders; Hairy Cell Leukemia; Chronic c7, Myelogeonus Leukemia; Hematological Malignancies; Hematological Disorders; Multiple t.o.) Myeloma; Kidney diseases and disorders;

Shonlein-Henoch purpura, Berger disease, celiac c7, disease, dermatitis herpetiformis, Chron disease;
Diabetes; Diabetes Insipidus; Diabetes mellitus;
Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia;
Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X; Kidney disorders;
oe Hyperinsulinemia; Hypoinsulinemia;
Immunological disorders (e.g. arthritis, asthma, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic -a 5 Protein:X Assay Protein:Z
immunodeficiency diseases, AIDS, rheumatoid arthritis, granulomatous disease, inflammatory bowl disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, T-cell mediated cytotoxicity, host-versus-graft disease, autoimmunity disorders, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjorgren's disease, scleroderma) C17 (cytokine- Inhibits glucose and/or Proliferation of kidney Kidney diseases and disorders; Shonlein-Henoch 2489, 2490 See Table 2, like protein C17) FFA uptake by mesangial cells can be purpura, Berger disease, celiac disease, SEQ ID
NO:Z 0 adipocytes; Induces assayed using techniques dermatitis herpetiformis, Chron disease; for particular proliferation of kidney described in J. Investig.
Diabetes; Diabetes Insipidus; Diabetes mellitus;
construct.
c7, mesangial cells; Med. (1998) Aug; Type 1 diabetes; Type 2 diabetes; Insulin Regulation of cytokine 46(6):297-302. resistance; Insulin deficiency; Hyperlipidemia;

production and antigen Hyperketonemia; Non-insulin dependent presentation Diabetes Mellitus (NIDDM);
Insulin-dependent 0 c7, Diabetes Mellitus (MDM); A Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X; Kidney disorders;
Hyperinsulinemia; Hypoinsulinemia;
Hematopoietic disorders; Immunological diseases and disorders; Developmental diseases and disorders; Hepatic diseases and disorders;
Cancer (particularly leukemia); Immunological disorders (e.g. arthritis, asthma, oe immunodeficiency diseases, AIDS, rheumatoid arthritis, granulomatous disease, inflammatory Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
bowl disease, sepsis, acne, neutropenia, neutrophilia, psoriasis, hypersensitivities, T-cell mediated cytotoxicity, host-versus-graft disease, autoimmunity disorders, demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjorgren's disease, sclero derma) HDPBQ71 Regulates production and Such assays that may be Blood disorders and infection (e.g., viral 2515, 2545 See Table 2, secretion of IFNgamma; used or routinely modified infections, tuberculosis, infections associated SEQ ID NO:Z
Activation of myeloid to test immunomodulatory with chronic granulomatosus disease and for particular cells and/or activity of polypeptides of malignant osteoporosis); Autoimmune disease construct. 0 hematopoietic cells the invention (including (e.g., rheumatoid arthritis, systemic lupus antibodies and agonists or erythematosis, multiple sclerosis);
antagonists of the invention) Immunodeficiency, boosting a T cell-mediated c7, include the assays disclosed immune response, and suppressing a T cell-in Miraglia et al., J mediated immune response;
Inflammation and 0 Biomolecular Screening inflammatory disorders;
Idiopathic pulmonary 0 4:193-204 (1999); Rowland fibrosis; Neoplastic diseases (e.g., leukemia, c7, et al., "Lymphocytes: a lymphoma, melanoma);
Neoplasms and cancers, practical approach" such as, for example, leukemia, lymphoma, Chapter 6:138-160 (2000); melanoma, and prostate, breast, lung, colon, Gonzalez et al., J Chin Lab pancreatic, esophageal, stomach, brain, liver and Anal 8(5):225-233 (1995); urinary cancer;. Benign dysproliferative disorders Billiau et al., Ann NY Acad and pre-neoplastic conditions, such as, for Sci 856:22-32 (1998); example, hyperplasia, metaplasia, and/or Boehm et al., Annu Rev dysplasia; Anemia;
Pancytopenia; Leukopenia;
Immunol 15:749-795 Thrombocytopenia;
Hodgkin's disease; Acute (1997), and Rheumatology lymphocytic anemia (ALL); Plasmacytomas;

(Oxford) 38(3):214-20 Multiple myeloma;
Burkitt's lymphoma;
(1999) Arthritis; AIDS;
Granulomatous disease;
Inflammatory bowel disease; Sepsis;
oe Neutropenia; Neutrophilia; Psoriasis;
Suppression of immune reactions to transplanted Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication.:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
organs and tissues; Hemophilia;
Hypercoagulation; Diabetes mellitus;
Endocarditis; Meningitis; Lyme Disease;
Asthma; Allergy Oscar (osteoclast- Regulator of osteoclast Assay to detect osteoclast Skeletal diseases and disorders; Musculoskeletal 2571, 2749 See Table 2, associated differentiation; regulator differentiation is described diseases and disorders; Bone fractures and/or SEQ ID NO:Z
receptor isoform- of innate and adaptive in J. Exp. Med. (2002) Jan breaks; Osteoporosis (postmenopausal, senile, or for particular 3) immune responses 21; 195(2):201-9. idiopathic juvenile);
Gout and/or pseudogout; construct.
Paget's disease; Osteoarthritis; Tumors and/or cancers of the bone (osteochondromas, benign chondromas, chondroblastomas, chondromyxoid fibromas, osteoid osteomas, giant cell tumors, multiple myelomas, osteosarcomas, c7, fibrosarcomas, malignant fibrous histiocytomas, \ chondrosarcomas, Ewing's tumors, and/or malignant lymphomas); Bone and joint infections (osteomyelitits and/or infectious arthritis);

c7, Charcot's joints; Heel spurs; Sever's disease;
Sport's injuries Tumstatin (T5, Inhibits angiogenesis;
A tumor cell proliferation Cancer; Solid Tumors; Melanoma; Malignant 2647, 2648, 2649, 2650, See Table 2, T7 or T8 peptide; Inhibits tumor growth;
assay is described in J. Biol. Melanoma; Renal Cancer (e.g., Renal Cell 2943, 2944, 3047, 3048 SEQ ID NO:Z
a3(IV)NC1) Inhibits protein synthesis Chem. (1997) 272:20395- Carcinoma);
Lung Cancer (e.g,. Non-Small Cell for particular 20401. Lung Cancer or Small Cell Lung Cancer) Colon construct.
Protein synthesis can be Cancer; Breast Cancer;
Liver Cancer; Prostate measured as described in Cancer; Bladder Cancer;
Gastric Cancer;
Science (2002) Jan 4; Sarcoma; AIDS-Related Kaposi's Sarcoma;
295(5552):140-3. Lymphoma; T Cell Lymphoma;
Cutaneous T-Cell Lymphoma; Non-Hodgkin's Lymphoma;

Brain Cancer; Glioma; Glioblastoma Multiforme;
Cervical Dysplasia; Leukemia; Preleukemia;
Bone Marrow Disorders; Bone Disorders; Hairy oe Cell Leukemia; Chronic Myelogeonus Leukemia;
Hematological Malignancies; Hematological Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay Protein:Z -a-, c, =
Disorders; Multiple Myeloma; Angiogenesis o --.1 CNTF (Ciliary Enhances myelin Regulation of myelin Neurological and neural diseases and disorders, 2724, 2725, 3171, 3172 See Table 2, neurotrophic formation; Reduces formation can be assayed as particularly diseases and disorders associated SEQ ID NO:Z
factor) photoreceptor described in J. Neurosci.
with myelin and demyelination, such as, for for particular degredation; Regulates (2002) Nov. 1; 22(21):9221- example, ALS, multiple sclerosis, Huntington's construct.
calcium currents 7. disease; Neuronal and spinal cord injuries;
Disorders of the eye, such as, for example, retinitis pigmentosa, blindness, color-blindness, macular degeneration.
n Somatostatin Inhibits growth hormone, Inhibition of growth Cancer; Metastatic carcinoid tumors; Vasoactive 2798, 2825, 2830, 2831, See Table 2, (Octreotide; glucagons and insulin;
hormone release in humans Intestinal Peptide secreting adenomas; Diarrhea 2902 SEQ ID NO:Z 0 I.) octreotide acetate; Suppresses LF response by somatostatin can be and Flushing; Prostatic disorders and cancers;
for particular a, -.3 Sandostating to GnRH; Decreases measured as described in J. Breast cancer; Gastrointestinal disorders and construct. H
CA
LARD) splanchnic blood flow; Clin. Endocrinol. Metab.
cancers; Cancers of the endocrine system; Head c7, u.) -i.
--.) Inhibits release of (1973) Oct; 37(4):632-4.
and neck paragangliomas; Liver disorders and I.) serotonin, gastrin, Inhibition of insulin cancers; Nasopharyngeal cancers; Thyroid 0 a, vasoactive intestinal secretion by somatostatin disorders and cancers;
Acromegaly; Carcinoid 0 peptide, secretin, motilin, can be measured as Syndrome; Gallbladder disorders, such as c7, I.) and pancreatic described in the Lancet gallbladder contractility diseases and abnormal H
polypeptide. (1973) Dec. 8; bile secretion;
Psoriasis; Diabetes; Diabetes 2(7841):1299-1301. Insipidus; Diabetes mellitus; Type 1 diabetes;
Type 2 diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia; Hyperketonemia;
Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A Condition Associated With Diabetes Iv Including, But Not Limited To Obesity, Heart n Disease, Hyperglycemia, infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune cp o Disorders; Obesity; Vascular Disorders;
t-.) Suppression of of Body Weight; Suppression of o oe Appetite; Syndrome X; Kidney disorders;
o 1-, Neurological disorders and diseases, including Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Alzheimers Disease, Parkinson's disease and dementia; Neuropsychotic disorders, including Bipolar affective disorder; Rheumatoid arthritis;
Hypertension; Intracranial hypertension;
Esophageal varices; Graves' disease; Seizures;
Epilepsy; Gastritis; Angiogenesis;
IL-22 (IL22, Stimulates glucose uptake IL-22 activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; 2901, 2903 See Table 2, interleulcin-22; in skeletal muscle cells; assayed in vitro using a [3-Diabetes mellitus; Type 1 diabetes; Type 2 SEQ ID NO:Z
IL17D, IL27) increases skeletal muscle Hi-glucose uptake assay. (J
diabetes; Insulin resistance; Insulin deficiency; for particular insulin sensitivity. Biol Chem 1999 Oct 22;
Hyperlipidemia; Hyperketonemia; Non-insulin construct.
274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin- 0 dependent Diabetes Mellitus (LDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or oo Ulcers; Metabolic Disorders; Immune Disorders;

Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X.
HCE1P80 Stimulates glucose uptake HCE1P80 activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; 2908, 3049, 3050, 3051, See Table 2, in; increases insulin assayed in vitro using a [3- Diabetes mellitus; Type 1 diabetes; Type 2 3052 SEQ ID NO:Z
sensitivity. 11]-glucose uptake assay. (J diabetes;
Insulin resistance; Insulin deficiency; for particular Biol Chem 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non-insulin construct.
274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
oo Syndrome X.
HDRMI82 Stimulates glucose HDRMI82 activity may be Hyperglycemia;
Diabetes; Diabetes Insipidus; 2909 See Table 2, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z -a 5 uptake; increases insulin assayed in vitro using a [3- Diabetes mellitus; Type 1 diabetes; Type 2 SEQ ID NO:Z =
o sensitivity. 11]-glucose uptake assay. (J diabetes;
Insulin resistance; Insulin deficiency; for particular 1--, Biol Chem 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non-insulin construct.
274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
n Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;

I.) Syndrome X.
a, -.3 HDALV07 Modulates insulin action Insulin activity may be Diabetes; Diabetes Insipidus; Diabetes mellitus;
3053, 3055, 3056 See Table 2, H
CA
c7, (adiponectin; assayed in vitro using a [3- Type 1 diabetes; Type 2 diabetes; Insulin SEQ ID NO:Z u.) t, gelatin-binding 1-1]-glucose uptake assay. (J resistance;
Insulin deficiency; Hyperlipidemia; for particular N) 28k protein Biol Chem 1999 Oct 22;
Hyperketonemia; Non-insulin dependent construct.

a, precurson; 274(43):30864-30873).
Diabetes Mellitus (NIDDM);
Insulin-dependent 1 c7, adipose most Diabetes Mellitus (IDDM); A Condition 1 I.) abundant gene Associated With Diabetes Including, But Not H
transcript; APM- Limited To Obesity, Heart Disease, 1; GBP28; Hyperglycemia, Infections, Retinopathy, And/Or ACRP30; Ulcers; Metabolic Disorders; Immune Disorders;
ADIPOQ) Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X; Hyperglycemia; Familial combined hyperlipidemia; Metabolic syndrome;
Iv Inflammatory disorders; Atherogenic disorders n 1-i C Peptide An insulin precursor C-peptide concentrations Diabetes; Diabetes Insipidus; Diabetes mellitus;
3088, 3149 See Table 2, involved in insulin can be measured using Type 1 diabetes; Type 2 diabetes; Insulin SEQ ID
NO:Z cp o regulation assays well known in the art, resistance;
Insulin deficiency; Hyperlipidemia; for particular t-.) such as as the one described in Hyperketonemia; Non-insulin dependent construct. a PNAS (1970) Sep; Diabetes Mellitus (NIDDM); Insulin-dependent vD
1--, 67(1):148-55 Diabetes Mellitus (IDDM); A Condition Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X; Hyperglycemia; Familial combined hyperlipidemia; Metabolic syndrome HCBOG68 Controls proliferation/ Activation of cAMP-Treatment of Obesity; treatment of Diabetes; 3106, 3270 See Table 2, (enteric differentiation or mediated transcription in suppression of body weight gain; suppression of SEQ ID NO:Z
adipokine; Fat metabolism/ adipocytes can be assayed appetite.
Hyperglycemia; Diabetes; Diabetes for particular 0 SID; proline rich physiology/pathology/ of using methods known in the Insipidus; Diabetes mellitus; Type 1 diabetes; construct.
acidic protein) adipocytes and adipose art (Berger et al., Gene Type 2 diabetes; Insulin resistance; Insulin tissue in response to 66:1-10 (1998); Cullen and deficiency;
Hyperlipidemia; Hyperketonemia; c7, dietary conditions. Maim, Methods in Enzymol Non-insulin dependent Diabetes Mellitus 216:362-368 (1992); (NIDDM); Insulin-dependent Diabetes Mellitus 0 Henthom et al., Proc Natl (IDDM); A Condition Associated With Diabetes Acad Sci USA 85:6342- Including, But Not Limited To Obesity, Heart c7, 6346 (1988); Reusch et al., Disease, Hyperglycemia, Infections, Retinopathy, Mol Cell Biol 20(3):1008- And/Or Ulcers; Metabolic Disorders; Immune 1020 (2000); and Klemm et Disorders; Obesity; Vascular Disorders;
al., J Biol Chem 273:917- Suppression of Body Weight; Suppression of 923 (1998)). Appetite; Syndrome X.
Other indications for antibodies and/or antagonists, include treatment of weight loss;
treatment of AIDS wasting; appetite stimulant;
treatment of cachexia.
PYY (Peptide Decreases appetite;
Appetite and food intake can Most preferred:
Treatment of Obesity; treatment 3108, 3109, 3281, 3117, See Table 2, 1-3 YY), including increases satiety; be can be measured by of Diabetes; suppression of body weight gain; 3118, 3282. SEQ ID
NO:Z
PYY3-36 decreases food intake. methods known in the art suppression of appetite. for particular (amino acid (Batterham et al. Nature Hyperglycemia; Diabetes; Diabetes Insipidus;
construct.
oe residues 31-64 of 2002; 418:650654) Diabetes mellitus; Type 1 diabetes; Type 2 full length PYY, diabetes; Insulin resistance; Insulin deficiency;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z -a-, amino acid Hyperlipidemia;
Hyperketonemia; Non-insulin =
o residues 3-36 of dependent Diabetes Mellitus (NIDDM); Insulin- 1--, mature PYY) dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, ' But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
n Syndrome X.
Other indications for antibodies, antagonists:

I.) treatment of weight loss; treatment of AIDS
a, -.3 H
wasting; appetite stimulant; treatment of u.) c7, cachexia.
u.) '1 WNT10b ,--, Inhibits adipogenesis. WNT10b activity can be Most preferred: Treatment of Obesity; 3141 See Table 2, I.) measured using suppression of body weight gain; suppression of SEQ ID NO:Z 0 a, adipogenesis inhibition appetite.
for particular 1 c7, assays (Ross et al., Science Other indications: Hyperglycemia; Diabetes;
construct. 1 I.) 2000; 289(5481):950-953 Diabetes Insipidus;
Diabetes mellitus; Type 1 H
diabetes; Type 2 diabetes; Insulin resistance;
Insulin deficiency; Hyperlipidemia;
Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM).
measured using assays Congestive Heart Failure; Myocardial Infarction. SEQ ID NO:Z
Iv known in the art, including for particular n ,-i cardiogenesis assays construct.
(Eisenberg et al., Dev Dyn o 1999 Sep;216(1):45-58).
Herstatin Inhibits cancer Herstatin activity can be Oncology; Cancer; Solid Tumors; Melanoma; 3143 See Table 2, ox proliferation, measured using cell Malignant Melanoma;
Renal Cancer (e.g., Renal SEQ ID NO:Z o 1--, proliferation assays known Cell Carcinoma); Lung Cancer (e.g,. Non-Small for particular Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z -a-, in the art (Doherty et al., Cell Lung Cancer or Small Cell Lung Cancer); construct.
o PNAS 1999; 96(19):10869- Colon Cancer; Breast Cancer; Liver Cancer;

1--, 10874. Prostate Cancer; Bladder Cancer; Gastric Cancer;
Sarcoma; AIDS-Related Kaposi's Sarcoma;
Lymphoma; T Cell Lymphoma; Cutaneous T-Cell Lymphoma; Non-Hodgkin's Lymphoma;
Brain Cancer; Glioma; Glioblastoma Multiforme;
Cervical Dysplasia; Leukemia; Preleukemia;
Hairy Cell Leukemia; Chronic Myelogeonus Leukemia; Hematological Malignancies;
n Hematological Disorders; Multiple Myeloma.

Adrenomedullin stimulates vasodilation; Vasodilation can be Treatment of Congestive Heart Failure; 3144 See Table 2, I.) a, -.3 promotes bone growth. measured using assays Hypertension; Myocardial Infarction; Septic SEQ ID NO:Z H
CA
known in the art (Ashton et Shock; Osteoporosis; Postmenopausal for particular c7, u.) tm al. Pharmacology 2000;
osteoporosis; Osteopenia. construct.
tv I.) 61(2):101-105. The a, promotion of bone growth can be measured using c7, I.) assays known in the art, H
such as the osteoblast proliferation assay (Cornish et al. Am J Physiol 1997 Dec;273(6 Pt 1):E1113-20).
Nogo Receptor Receptor for the axon The promotion of axon Treatment of Central Nervous System Damage; 3184, 3185 See Table 2, growth inhibitor, Nogo. regeneration and growth can Spinal Cord Injury;
Peripheral Nerve Damage; SEQ ID NO:Z
be measured using assays Neurodegenerative Diseases; Parkinson's for particular Iv known in the art (Fournier et Disease; Alzheimer's Disease; Huntington's construct. n al. Nature 2001; Disease; Amyotrophic Lateral Sclerosis; 1-3 409(6818):341-346). Progressive Supranuclear Palsy; Creutzfeld-cp o Jacob Disease; Motor Neuron Disease.
t-.) CART (Cocaine- Inhibits food intact and Appetite and food intake can Most preferred: Treatment of Obesity; 3232 See Table 2, o oe 1--, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z -a-, Regulated (Batterham et al. Nature Other indications: Hyperglycemia; Diabetes;
construct. =
o Transcript) 2002; 418:650654) Diabetes Insipidus;
Diabetes mellitus; Type 1 1--, diabetes; Type 2 diabetes; Insulin resistance;
Insulin deficiency; Hyperlipidemia;
Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM).
RegIV (Colon Stimulates glucose RegIV activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; 2910. See Table 2, Specific Gene; uptake; increases insulin assayed in vitro using a [3-Diabetes mellitus; Type 1 diabetes; Type 2 SEQ ID NO:Z
n Colon Specific sensitivity. HI-glucose uptake assay. (J diabetes;
Insulin resistance; Insulin deficiency; for particular Protein) Biol Chem 1999 Oct 22;
Hyperlipidemia; Hyperketonemia; Non-insulin construct. 0 I.) 274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin- a, -.3 dependent Diabetes Mellitus (IDDM); A
H
CA
c7, Condition Associated With Diabetes Including, co cal But Not Limited To Obesity, Heart Disease, I.) Hyperglycemia, Infections, Retinopathy, And/Or a, Ulcers; Metabolic Disorders; Immune Disorders;

c7, Obesity; Vascular Disorders; Suppression of I.) Body Weight; Suppression of Appetite;
H
Syndrome X.
Cosyntropin Synthetic corticotropin; The activity of cosyntropin Endocrine; Addison's disease; Cushing's SEQ ID:
(Cortrosyn) stimulates the release of can be assessed in vivo by syndrome;
pituitary dysfunction; acute adrenal NO :2198 (CAS-16960-16- cortisol. measuring serum cortisol crisis 0) levels. (Frank et al. J. Am.
Vet. Med. Assoc. 1998 212(10):1569-71).
Iv Pexiganan Disrupts bacterial Pexiganan acetate activity Treatment of Infectious Diseases; Treatment of SEQ ID NO: n Acetate membranes, can be assessed using in Bacterial Infections. 2199 (CAS-172820-23- vitro antibacterial assays cp o 4) known in the art. (Zasloff et t-.) al., Antimicrobial Antimicrobial Agents a and Chemotherapy 1999, vD
1--, 43:782-788).
_ Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z -a-, _ =
Pramlintide Slows gastric emptying; Appetite and food intake can Treatment of Obesity; treatment of Diabetes; SEQ ID NO: =

(Amylin) decreases food intake, be can be measured by suppression of body weight gain; suppression of 2200 1--, (CAS-151126-32- methods known in the art appetite;
treatment of endocrine disorders;
8) (Batterham et al. Nature Hyperglycemia;
Diabetes; Diabetes Insipidus;
2002; 418:650654) Diabetes mellitus; Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency;
Hyperlipidemia; Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
n Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, I.) Hyperglycemia, Infections, Retinopathy, And/Or a, -.3 Ulcers; Metabolic Disorders; Immune Disorders;
H
CA
c7, Obesity; Vascular Disorders; Suppression of u.) vi Body Weight; Suppression of Appetite; I.) -4.

Syndrome X.

a, Other indications for antibodies, antagonists:

c7, treatment of weight loss; treatment of AIDS

I.) wasting; appetite stimulant; treatment of H
cachexia.
metabolism; elevates oyariectomized rat model of Osteoclastogenesis; osteolysis; osteomyelitis;
blood calcium level; osteoporosis: LUBMB Life osteonecrosis;
periodontal bone loss;
stimulates the activity of 2000 Feb;49(2):131-5 osteoarthritis;
rheumatoid arthritis; osteopetrosis;
Iv osteocytes; enhances periodontal, lytic, or metastatic bone disease; n ,-i absorption of Ca+/Pi from osteoclast differentiation inhibition; bone small intestine into blood; disorders; bone healing and regeneration. cp o promotes reabsorption of Ca+ and inhibits Pi by a kidney tubules.
o 1--, _ Table 1 -Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Terlipressin Analog of vasopressin; Terlipressin activity can be Variceal hemorrhage; cirrhosis; portal SEQ ID NO:
(Iriglycyl lycine induces vasoconstriction. measured using assays of hypertension; hepatorenal syndrome; Blood- 2202 vasopressin) vasoconstriction, such as the related disorders (CAS-14636-12- isolated arterial ring 5) preparation. (Landstrom et al., Hum Reprod 1999 Jan;14(1):151-5).
Ularitide Stimulates natriuresis, Ularifide activity can be Excretory disorders; Acute renal failure; asthma;
SEQ ID NO:
(CAS-118812-69- diuresis, and vasodilation. assessed by measuring congestive heart failure; hypertension; pulmonary 2203 o 4) cGMP accumulation in rat hypertension;
cardiovascular disorders renal cells. (Valentin et al., 1..) Hypertension 1993 0.

Apr;21(4):432-8).
w 0, Aprotinin Serine protease inhibitor; Inhibition of thrombin-Inhibition of fibrinolysis; reduction of blood loss SEQ ID NO: w (Trasylol)TM
attenuates Systemic induced platelet aggregation during surgery;
Treatment of Inflammation and 2204 "

(CAS-9087-70-1; Inflammatory Response, can be measured using Immune Disorders.

CAS-11061-94-2; ffbrinolysis and thrombin- methods known in the art.

CAS-12407-79-3) induced platelet (Poullis et al., J Thorac ko 1..) aggregation. Cardiovasc Surg 2000 Aug;1200:370-8).
Aspartocin Antibacteria Aspartocin activity can be Treatment of Infectious Diseases; treatment of SEQ ID NO:
(CAS-4117-65-1; assessed using in vitro bacterial infections. 2205 CAS-1402-89-7) antibacterial assays known .
in the art. (Zasloff et al., Antimicrobial Agents and Chemotherapy 1999, 43:782-788).
Calcitonin Regulates levels of Hypocalcemic Rat Bioassay, Musculoskeletal;
Osteroporosis; Paget's disease; SEQ ID NO:
(Calcimar) calcium and phosphate in bone resorbing assay and the hypercalcemia; 2206 (CAS-21215-62- serum; causes a reduction pit assay, CT receptor Bone Disorders; Fracture prevention; Malignant 3) in serum calcium¨an binding assay, CAMP
hypercalcemia; Osteopenia, Osteoclastogenesis;
effect opposite to that of stimulation assay: J Bone osteolysis;
osteomyelitis; osteonecrosis;
human parathyroid Miner Res 1999 periodontal bone loss;
osteoarthritis; rheumatoid Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z
hormone. Aug;14(8):1425-31 arthritis;
osteopetrosis; periodontal, lytic, or =
o metastatic bone disease; osteoclast differentiation inhibition; bone disorders; bone healing and regeneration.
Carperitide Stimulates natriuresis, Carperitide activity can be Treatment of Heart Failure; Cardiovascular SEQ ID NO:
(HANP; diuresis, and vasodilation. assessed in vitro by disorders; Respiratory disorders; Acute 2207 recombinant measuring cGMP respiratory distress syndrome.
human atrial accumulation in a number of natriuretic cell lines, including PC12 -n peptide) cells and cultured human (CAS-89213-87- glomerular cells. (Medvede I.) 6) et al., Life Sci 2001 Aug a, -.3 31;69(15):1783-90; Green H
CA
c7, et al., J Am Soc Nephrol u.) 1994 Oct;5(4):1091-8).
N) <a Desirudin Inhibits thrombin; inhibits Desirudin activity can be Blood-related disorder; Thrombosis; SEQ ID NO:

a, (recombinant blood clotting, assessed using blood thrombocytopenia; hemorrhages. 2208 0 c7, hirudin; Revasc) clotting assays known in the I.) (CAS-120993-53- art, such as in vitro platelet H
5) aggragation assays. (Glusa, Haemostasis 1991;21 Suppl 1:116-20).
Emoctalcin proinflammatory cytolcine Treatment of Inflammation, Immune disorders, SEQ ID NO:
(interleukin 8) RSV infection.

(CAS-142298-00-8) n Felypressin Derivative of Felypressin vasoconstriction Treatment of pain; to induce local anesthesia. SEQ ID NO:
cp (CAS-56-59-7) Vasopressin; Stimulates activity can be measured vasoconstriction; Induces using assays of local anesthesia. anesthesia. vasoconstriction, such as the a vD
1-, isolated arterial ring Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a Protein:X Assay Protein:Z
preparation. (Landstrom et al., Hum Reprod 1999 Jan;14(1):151-5).
Glucagon Induces hyperglycemia. Glucagon activity may be Hypoglycemia;
Diabetes; Diabetes Insipidus; SEQ ID NO:
(CAS-16941-32- assayed in vitro using a [3- Diabetes mellitus; Type 1 diabetes; Type 2 2211 5) HI-glucose uptake assay. (J diabetes; Insulin resistance; Insulin deficiency;
Biol Chem 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non-insulin 274(43):30864-30873). dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
LT, Obesity; Vascular Disorders; Suppression of LA
Body Weight; Suppression of Appetite;

Syndrome X; Endocrine disorders.

Nagrestipen Inflammation; Immune SEQ ID NO: 0 (CAS-166089-33-2212 LT, Pentigetide Respiratory; Allergy;
Immune SEQ ID NO:
(Pentyde) (CAS-62087-72-3) Proinsulin Stimulates glucose uptake Insulin activity may be Hyperglycemia; Diabetes; Diabetes Insipidus; SEQ
ID NO: 1-3 (CAS-67422-14- and promotes assayed in vitro using a [3- Diabetes mellitus; Type 1 diabetes; Type 2 2214 cA
4) glycogenesis and H]-glucose uptake assay. (J diabetes; Insulin resistance; Insulin deficiency;
lipogenesis. Biol Chem 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non-insulin oe 274(43):30864-30873). dependent Diabetes Mellitus (NMDM); Insulin-dependent Diabetes Mellitus (IDDM); A

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite;
Syndrome X.
Becaplermin Promotes wound healing. Becaplermin activity can be Stimulate Epithelial Cell Proliferation; Stimulate SEQ ID NO:
(Regranex; assessed using animal Basal Keratinocytes; Promote Wound Healing; 2215 recombinant wound healing models Stimulate Hair Follicle Production; Healing Of PDGF-BB) known in the art. (Saba et Dermal Wounds.
Wound Healing; Eye Tissue 0 (CAS-165101-51- al., Ann Plast Surg 2002 Wounds, Dental Tissue Wounds, Oral Cavity 9) Jul;49(1):62-6). Wounds, Diabetic Ulcers, Dermal Ulcers, Cubitus Ulcers, Arterial Ulcers, Venous Stasis c7, oo Ulcers, Burns Resulting From Heat Exposure Or Chemicals, or Other Abnormal Wound Healing Conditions such as Uremia, Malnutrition, Vitamin Deficiencies or Complications c7, Associated With Systemic Treatment With Steroids, Radiation Therapy or Antineoplastic Drugs or Antimetabolites; Promote Dermal Reestablishment Subsequent To Dermal Loss;
Increase the Adherence Of Skin Grafts To A
Wound Bed; Stimulate Re-Epithelialization from The Wound Bed; To Promote Skin Strength;
Improve The Appearance Of Aged Skin;
Proliferate Hepatocytes, Lung, Breast, Pancreas, Stomach, Bladder, Small Intestine, Large Intestine; Sebocytes, Hair Follicles, Type II
Pneumocytes, Mucin-Producing Goblet Cells, or Other Epithelial Cells, Endothelial Cells, oe Keratinocytes, or Basal Keratinocytes (and Their Progenitors) Contained Within The Skin, Lung, Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
Liver, Bladder, Eye, Salivary Glands, or Gastrointestinal Tract; Reduce The Side Effects Of Gut Toxicity That Result From Radiation, Chemotherapy Treatments Or Viral Infections;
Cytoprotector, especially of the Small Intestine Mucosa or Bladder; Mucositis (Mouth Ulcers);
Regeneration Of Skin; Full and/or Partial Thickness Skin Defects, including Burns, (e.g., Repopulation Of Hair Follicles, Sweat Glands, And Sebaceous Glands); Psoriasis;
Epidermolysis Bullosa; Blisters; Gastric and/or Doudenal Ulcers; Reduce Scarring;
Inflamamatory Bowel Diseases; Crohn's Disease;
Ulcerative Colitis; Gut Toxicity; Lung Damage;
c7, Repair Of Alveoli And/or Brochiolar Epithelium;

Acute Or Chronic Lung Damage; Emphysema, ARDS; Inhalation Injuries; Hyaline Membrane Diseases; Infant Respiratory Distress Syndrome;
c7, Bronchopulmonary Displasia In Premature Fa Infants; Fulminant Liver Failure; Cirrhosis, Liver Damage caused by Viral Hepatitis and/or Toxic Substances; Diabetes Mellitus; Inflammation;
Cancer; Digestive disorders.
Ghrelin Stimulates release of Appetite and food intake can Endocrine; loss of body weight; loss of body SEQ ID NO:
(Genbank growth hormone from be can be measured by weight associated with cancer or anorexia 2216 Accession No. anterior pituitary. methods known in the art nervosa; loss of appetite; excessive appetite;
AB029434) Stimulates appetite and (Batterham et al. Nature body weight gain; Obesity; Diabetes;
reduces fat burning. 2002; 418:650654) Acromegaly; Growth failure; Growth hormone 1-3 deficiency; Growth failure and growth retardation Prader-Willi syndrome in children 2 years or older; Growth deficiencies; Growth failure oe associated with chronic renal insufficiency;
Postmenopausal osteoporosis; burns; cachexia;
_ Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z -a-, c, =
cancer cachexia; dwarfism; metabolic disorders;
o obesity; renal failure; Turner's Syndrome, pediatric and adult; fibromyalgia; fracture = treatment; frailty, AIDS wasting Ghrelin -binding Inhibits growth hormone Appetite and food intake can Endocrine; Obesity; Diabetes; body weight gain;
antibody release in response to be can be measured by excessive appetite; loss of appetite; loss of body including Ghrelin; inhibits increase methods known in the art weight.
antibody in appetite. (Batterham et al. Nature fragment, or 2002; 418:650654) n dominant-negative form of I.) Ghrelin receptor a, -.3 NOGO-66 Neurodegenerative disorders; spinal cord injury; SEQ ID NO: H
CA
peptide fragment neuronal injury; brain trauma; stroke; multiple 2217 c7, u.) (Genbank sclerosis;
demyelinating disorders; neural activity I.) Accession No. and neurological diseases; neural cell (e.g., 0 a, NP_008939 neuron, glial cell, and schwann cell) regeneration 0 (amino acids 62- and/or growth c7, , 101)) I.) H
Gastric inhibitory Increases nutrient uptake Nutrient uptake and Most preferred: loss of body weight, AIDS SEQ
ID NO:
polypeptide and tryglyceride tryglyceride accumulation wasting, cachexia, loss of apetite. Other: 2218 (GIP), including accumulation in can be measured by methods Obesity;
Diabetes; insulin resistance; body GIP fragments adipocytes, which leads to desribed in Miyawaki et al., weight gain; excessive appetite.
(Genbank obesity and insulin Nat. Medicine, 2002, Vol Accession No. resistance. 8(7):738-742.
NM 004123) Iv n ,-i Gastric inhibitory Increased use of fat as Fat utilization as an energy Obesity; Diabetes; Insulin resistance; body cp polypeptide predominant energy source can be measured as weight gain.
o antibody, or source; decreased described in Miyawalci et o antibody accumulation of fat in al., Nat. Medicine, 2002, oe vD
fragments adipocytes. Vol 8(7):738-742.

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z -a-, c, Gastric inhibitory Increased use of fat as Fat utilization as an energy Most preferred:
Obesity; Diabetes; body weight SEQ ID NO: o o peptide receptor predominant energy source can be measured as gain; excessive appetite;
insulin resistance. 2219 1--, or receptor source; decreased described in Miyawaki et Other: loss of body weight, AIDS wasting, loss of fragments or accumulation of fat in al., Nat. Medicine, 2002, appetite.
variants including adipocytes. Vol 8(7):738-742.
soluble fragments or variants (Genbank Accession Number n NM 000164) I.) POMC Activity of POMC- Preferred: resistance to stress; anti-inflammatory SEQ ID NO: a, -.3 (proopiomelanoco derived fragments are activity; analgesic activity; increased skin 2220 H
u.) rtin), including diverse, and well-known pigmentation; increased protein catabolism; c7, u.) fragments or in the art. increased gluconeogenesis;
obesity; diabetes. I.) variants (such as, Other: decreased protein catabolism, decreased 0 a, for example, See, for example, Hadley skin pigmentation, Addison's disease, Cushing's 0 alpha-melanocyte et al., Ann N Y Acad Sci syndrome stimulating 1999 Oct 20;885:1-21;
I.) Fa hormone, aMSH, Dores, Prog Clin Biol Res gamma 1990;342:22-7; Blalock, melanocyte Ann NY Acad Sci. 1999 stimulating Oct 20;885:161-72).
.
hormone, yMSH, beta-melanocyte stimulating Iv hormone, f3MSH, n adrenocorticotrop in, ACTH, beta-cp o endorphin, met-t-.) enkephalin) o oe (Genbank vD
1--, Accession No.

Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
Protein:X Assay Protein:Z -a-, NM 000930) =
o HP 467, HP228 See US Patent No. See US Patent No. Resistance to stress;
anti-inflammatory activity; SEQ ID NO: --.1 1--, (US Patent No. 6,350,430 6,350,430 analgesic activity;
increased skin pigmentation; 2221 6,350,430) increased protein catabolism; increased gluconeogenesis.
NDP See US Patent No. See US Patent No. Resistance to stress; anti-inflammatory activity; SEQ ID NO:
(US Patent No. 6,350,430 6,350,430 analgesic activity;
increased skin pigmentation; 2222 6,350,430) increased protein catabolism; increased n gluconeogenesis.

I.) a, Interleulcin-21 Immunomodulator; IL-21 activity can be Autoimmune disorders; Inflammatory disorders; 3298 SEQ ID NO:
H
(IL-21) inhibits interferon gamma assessed by measuring Treatment of Psoriasis; Rheumatoid Arthritis;
2177 u.) c7, u.) cA production by Thl cells. interferon gamma Inflammatory bowel disease.
Iv I.) production in Thl cells.

a, (Wurster et al., : J Exp Med 2002 Oct 7;196(7):969-77) m Interleukin-4 Immunomodulator; IL-4 activity can be assessed Treatment of Psoriasis; Autoimmune disorders; 3307 SEQ ID NO: "
H
(IL-4) promotes the by measuring Thl / Th2 Rheumatoid Arthritis; Inflammatory bowel 2178 differentiation of T cells cytokine responses of disease; Inflammatory disorders.
into Th2 phenotype. isolated spleen cells in vitro.
(Waltz et al., Horm Metab Res 2002 Oct;34(10):561-9).
Osteoclast Inhibits osteoclast Osteoclast Inhibitory Lectin Treatment of Bone Disorders;
Osteoporosis; 3312 SEQ ID NO:
Inhibitory Lectin formation. activity can be assessed Fracture prevention; Hypercalcemia; Malignant n (OCIL) using osteoclast formation hypercalcemia;
Paget's disease; Osteopenia, 1-3 assays known in the art. Osteoclastogenesis;
osteolysis; osteomyelitis;
cp (Zhou et al., J Biol Chem osteonecrosis;
periodontal bone loss; o n.) 2002 Dec osteoarthritis;
rheumatoid arthritis; osteopetrosis;

o 13;277(50):48808-15) periodontal, lytic, or metastatic bone disease; oe o 1--, osteoclast differentiation inhibition; bone healing Table 1 C
Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
and regeneration.

UJ
UJ
ON
tJF) oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader o No. ID Vector ID ID ID ID ID Sequence F, NO:Y NO: NO:Z NO: NO:B
X
A
1 1520 pC4:HSA/TR6.V30- Amino acids V30 to H300 of TR6 pC4 H300 (fragment shown as Vito H271 of SEQ
ID NO :433) fused downstream of HSA.
2 1537 pYPG:HSA.TR6c0V30- Amino acids V30 to E294 of TR6 pYPGaf E294 (fragment shown as Vito E265 of SEQ
ID NO:434) fused downstream of HSA.
DNA encoding TR6 has been codon optimized.
\ 3 1545 pYPG:HSA.TR6c0V30- Amino acids V30 to L288 of TR6 pYPGaf (5) L288 (fragment shown as Vito L259 of SEQ
ID NO:435) fused downstream of HSA.

DNA encoding TR6 has been codon optimized.
(5) 4 1546 pYPG:HSA.TR6c0V30- Amino acids V30 to R284 of TR6 pYPGaf R284 (fragment shown as Vito R255 of SEQ
ID NO:436) fused downstream of HSA.
DNA encoding TR6 has been codon optimized.
1568 pSAC35:HSA-yTR6 TR6 fused downstream of HSA. DNA pSAC35 221 5 437 657 658 HSA/kex2 encoding TR6 has been codon optimized.
6 1570 pSAC35:TR6-HSA Mature TR6 fused downstream of the pSAC35 222 6 438 659 660 HSA/kex2 HSA/kex2 leader and upstream of the mature HSA.
7 1622 pC4:synTR6.M1- Synthetic TR6 fused upstream of mature pC4 223 7 439 661 662 Native TR6 H300.HSA HSA, with 2 extra amino acids between oe the TR6 and HSA portions.

=
=
Table 2 0 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence g;
NO:Y NO: NO:Z NO: NO:B
X
A
8 1623 pC4:HSA.synTR6.V30- Synthetic mature TR6 fused downstream pC4 H300 of FL HSA. Last amino acid HSA
sequence is missing at BSU36I site.
9 1642 pSAC35:GCSF.T31- Mature GCSF cloned downstream of the pSAC35 225 9 441 665 666 HSA/kex2 P204.HSA HSA/kex2 leader and upstream of the mature HSA
1643 pSAC35:HSA.GCSF.T3 Mature GCSF cloned downstream of the pSAC35 226 10 442 667 668 HSA/kex2 1-P204 mature HSA and HSA/kex2 leader sequence.
8; 11 1645 pSAC35:yTR6(N173Q). Mutant mature TR6 cloned upstream of pSAC35 227 11 443 669 670 HSA/kex2 (5) HSA mature HSA and downstream of the HSA/kex2 leader sequence.

12 1657 pC4.HSA:DeCAF.A23- Amino acids A23 to D233 of DeCAF pC4 (5) D233 fused downstream of full length HSA.
13 1680 pYPG:HSA.BLyS.A134 Amino acids A134 to L285 of BLyS fused pYPGaf -L285 downstream of FL HSA. Two extra amino acids (Leu, Glu) have been added between the therapeutic protein and HSA
portions.
14 1681 pYPG.HSA.MPIF.D45- Amino acids D45 to N120 of MPIF fused pYPGaf N120 downstream of FL HSA. Two additional 1-d amino acids (L and E) have been added between HSA and MPIF.
1697 pSAC35:HSA.GM- Amino acids A18 to E144 of GM-CSF pSAC35 CSF.A18-E144 fused downstream of FL HSA.
oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader e.) No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B
=

X
A 1.., 16 1699 pSAC35:GM-CSF.A18- Amino acids A18 to E144 of GM-CSF
pSAC35 232 16 448 679 680 HSA/kex2 E144:HSA fused upstream of mature HSA and downstream of HSA/kex2 leader.
17 1700 pSAC35:HSA- Mutant TR6 fused downstream of mature pSAC35 233 17 449 681 682 HSA/kex2 yTR6(N173Q) HSA with HSA/kex2 leader sequence.
18 1702 pYPG:HSA.ek.TR6coV Amino acids V30 to L288 of TR6 pYPGaf n 30-L288 (fragment shown as Vito L259 of SEQ
ID NO:450) fused downstream of FL

I.) a, HSA with an enterokinase site in between.
H
Cl 0\ DNA encoding TR6 has been codon co (5) co optimized.
I.) 19 1703 pYPG:HSA.ek.TR6coV Amino acids V30 to R284 of TR6 pYPGaf 30-R284 (fragment shown as Vito R255 of SEQ
a, ID NO:451) fused downstream of HSA
(5) with an enterokinase site in between.
I.) H
DNA encoding TR6 has been codon optimized.
20 1704 pYPG:HSA.TR6.V30- Amino acids V30 to E294 of TR6 fused pYPGaf E294 downstream of HSA. Two additional amino acids (Leu, Glu) are in between HSA and TR6.
21 1746 pYPG:HSA.ek.KDI.L28 Amino acids L28 to 1(207 of KDI fused pYPGaf 237 21 453 689 690 HSA 1-d n -1(207 downstream of HSA with an enterokinase site in between.
cp 22 1747 pSAC35.HSA.hGHRF. Amino acids Y32 to L75 of hGHRF fused pSAC35 238 22 454 691 692 HSA c' t..) Y32-L75 downstream of HSA.

o oe o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader E' No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
23 1748 pSAC35.hGHRF.Y32- Amino acids Y32 to L75 of hGHRF (see pSAC35 239 23 455 693 694 HSA/kex2 L75.HSA also SEQ IDNO:454) fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
24 1749 pSAC35:HSA.PTH.S1- FL HSA fused upstream of amino acids pSAC35 240 24 456 695 696 HSA
F3 S1-F34 of PTH
25 1750 pSAC35:PTH.S1- Amino acids 1-34 of PTH fused upstream pSAC35 241 25 457 697 698 HSA/kex2 F34.HSA of mature HSA and downstream of HSA/kex2 leader sequence.
26 1757 pSAC35:IL2.A21- Mature human IL-2 with a single amino pSAC35 242 26 458 699 700 HSA/kex2 (5) T153.145C/S.HSA acid mutation (C to S at position 145) cloned downstream of the HSA/KEX2 leader and upstream of mature HSA

27 1758 pSAC35:HSA.EL2.A21- Mature human IL-2 with a single amino pSAC35 243 27 459 701 702 HSA/kex2 (5) T153.145C/S acid mutation (C to S at position 145) cloned downstream of HSA with HSA/kex2 leader sequence.
28 1772 pSAC:EPOco.A28- Amino acids A28-D192 of EPO variant pSAC35 244 28 460 703 704 HSA/kex2 D192.HSA (where glycine at amino acid 140 has been replaced with an arginine) fused upstream of mature HSA and downstream of HSA/kex2 leader sequence. DNA
1-d encoding EPO has been codon optimized.
oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence cl;
NO:Y NO: NO:Z NO: NO:B
X
A
29 1774 pSAC:HSA.EPOco.A28 Amino acids A28-D192 of EPO variant pSAC35 245 29 461 705 706 HSA/kex2 -D192. (where glycine at amino acid 140 has been replaced with an arginine) fused downstream of HSA with HSA/kex2 leader sequence. DNA encoding EPO has been codon optimized.
30 1777 pSAC35:TNFR2.L23- Mature TNFR2 fused downstream of the pSAC35 246 30 462 707 708 HSA/kex2 D257.HSA HSA/kex2 signal and upstream of mature HSA.
00 31 1778 pSAC35:lFN13.M22- Residues M22-N187 of full-length TINb pSAC35 247 31 463 709 710 HSA/kex2 (5) N187:HSA (shown as M1 to N166 of SEQ ID

NO:463) fused upstream of mature HSA

and downstream of HSA/kex2 leader (5) sequence.
32 1779 pSAC35:HSA:IFN13.M2 Residues M22-N187 of full-length IFNb pSAC35 248 32 464 HSA/kex2 2-N187 (shown as M1 to N166 of SEQ ID
NO :464) fused downstream of HSA with HSA/kex2 leader sequence.
33 1781 pSAC:EPOcoA28- Amino acids A28-D192 of EPO variant pSAC35 249 33 465 711 712 HSA/kex2 D192.HSA (where glycine at amino acid 140 has been 51N/S,65N/S,110N/s replaced with an arginine) fused upstream 1-d of mature HSA and downstream of HSA/kex2 leader sequence. Glycosylation sites at amino acid 51, 65, 110 are mutated from N to S residue. DNA
encoding EPO has been codon optimized.
oe Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader st,' o No. ID Vector ID ID ID ID ID Sequence E
=
NO:Y NO: NO:Z NO: NO:B

1.., X
A
34 1783 pSAC:HSA.EPOcoA28- Amino acids A28-D192 of EPO variant pSAC35 250 34 466 713 714 HSA/kex2 D192.51N/S,65N/S,110 (where glycine at amino acid 140 has been N/s replaced with an arginine) fused , downstream of HSA with HSA/kex2 leader sequence. Glycosylation sites at amino acids 51, 65, 110 are mutated from n N to S residue. DNA encoding EPO has been codon optimized.
I.) a, 35 1784 pSAC35:HSA.TNFR2.L Mature TNFR2 fused downstream of FL pSAC35 H
23-D257 HSA.
co (5) co 36 1785 pSAC35:KGF2 28.A63 Amino acids A63 to S208 of KGF2 fused pSAC35 252 36 468 717 718 HSA/kex2 I.) -S208:HSA upstream of mature HSA and downstream a, ' of the HSA/kex2 signal peptide.

37 1786 pSAC35:HSA.KGF2{D Amino acids A63 to S208 of KGF2 fused pSAC35 253 37 469 719 720 HSA (5) I.) }28.A63-S208 downstream of HSA.
H
38 1788 pSAC35:HSA.TR2.P37- Amino acids P37 to A192 of TR2 fused pSAC35 254 38 470 721 722 HSA/kex2 A192 downstream of HSA with HSA/kex2 leader sequence.
39 1793 pSAC35:HSA.EPO.A28 Amino acids A28-D192 of EPO variant pSAC35 255 39 471 HSA/kex2 -D192 (where glycine at amino acid 140 has been (N51A,N65A,N110A) replaced with an arginine; see, for example, SEQ ID NO :499) fused Iv n downstream of HSA with HSA/kex2 leader sequence. Glycosylation sites at cp o amino acids 51, 65, 110 are mutated from t..) N to A residue.
o oe o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader w No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
40 1794 pSAC35:HSA.EPO.A28 Amino acids A28-D192 of the EPO pSAC35 256 40 472 HSA/kex2 -D192 variant (where glycine at amino acid 140 has been replaced with an arginine; see, for example, SEQ ID NO:499) fused downstream of HSA with HSA/kex2 leader sequence.
41 1809 pSAC35.MDC.G25- Amino acids P26 to Q93 of MDC with an pSAC35 257 41 473 723 724 HSA/kex2 Q93 .HSA N-terminal methionine, fused downstream of the HSA/kex2 leader and upstream of mature HSA.
(5) 42 1812 pSAC35:IL2.A21- Amino acids A21 to T153 of 1L-2 fused pSAC35 258 42 474 725 726 HSA/kex2 T153.HSA downstream of the HSA/kex2 leader and upstream of mature HSA.

43 1813 pSAC35:HSA.IL2.A21- Amino acids A21 to T153 of IL-2 fused pSAC35 259 43 475 727 728 HSA/kex2 (5) T153 downstream of HSA with HSA/kex2 leader sequence.
44 1821 pSAC35:scFv116A01.H BLyS antibody fused upstream of mature pSAC35 260 44 476 729 730 Modified SA HSA which lacks the first 8 amino acids HSA/kex2, and downstream from the HSA/kex2 lacking the signal sequence which lacks the last two last two amino acids.
amino acids 1-d 45 1830 pSAC35:HSA.KEX2.H Amino acids L19-Q300 of HAGDG59 pSAC35 261 45 477 731 732 HSA/kex2 AGDG59.L19-Q300 fused downstream of the HSA/kex2 signal, mature HSA and KEX2 cleavage site.
oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader E' No. ID Vector ID ID ID ID ID Sequence o NO:Y NO: NO:Z NO: NO:B
=

X
A 1.., 46 1831 pSAC35:HAGDG59.L1 HSA/kex2 signal peptide followed by pSAC35 262 46 478 733 734 HSA/kex2 9-Q300.HSA amino acids L19-Q300 of HAGDG59 followed by mature HSA.
47 1833 pSAC35:humancalcitoni Human Calcitonin (amino acids C98-pSAC35 263 47 479 735 736 HSA/kex2 n. C 1-033 :HSA G130 of SEQ ID NO:479) fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
n 48 1834 pSAC35:HSA.humancal Human Calcitonin (amino acids C98-pSAC35 264 48 480 737 738 HSA 0 I.) citonin.C1-G33 G130 of SEQ ID NO:480) fused a, -,1 =====1H
,¨ downstream of FL HSA.
co (5) 49 1835 pSAC35:salmoncalcitoni Salmon Calcitonin amino acids C1-G33 pSAC35 265 49 481 739 740 HSA/kex2 co I.) n.C1-G33:HSA fused upstream of mature HSA and . downstream of HSA/kex2 leader a, , sequence.
(5) 50 1836 pSAC35:HSA.salmoncal Salmon Calcitonin amino acids Cl-G33 pSAC35 266 50 482 741 742 HSA "
H
citonin.C1-G33 fused downstream of HSA.
51 1853 pSAC35:PTH(1- Amino acids 1 to 34 of PTH fused pSAC35 267 51 483 743 744 HSA/kex2 34)N26.HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence. Amino acid 1(26 of PTH mutated to N26.
52 1854 pSAC35:HSA.PTH(1- Amino acids 1 to 34 of PTH fused pSAC35 268 52 484 745 746 HSA
34)N26 downstream of HSA. Amino acid 1(26 of 1-d n PTH mutated to N26.
53 1862 pSAC35:HSA.GnRH.Q Amino acids Q24-G33 of human pSAC35 269 53 485 747 748 HSA/kex2 c7, 24-033 gonadotropin releasing hormone fused o t..) .1-.
downstream of HSA with HSA/kex2 o oe leader sequence.
vD


Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence E
=
NO:Y NO: NO:Z NO: NO:B

1.., X
A
54 1863 pSAC35:GnRHQ24- Amino acids Q24-G33 of human pSAC35 270 54 486 749 750 HSA/kex2 G33.HSA gonadotropin releasing hormone fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
55 1866 pSAC35:teprotide.HSA Teprotide fused upstream of mature HSA. pSAC35 271 55 487 751 752 n 56 1867 pSAC35:HSA.teprotide. Teprotide fused downstream of FL
HSA. pSAC35 272 56 488 753 754 HSA

57 1889 pC4:HSA.PTH.S1-F34 PTH(1-34) fused downstream of HSA.
pC4 273 57 489 755 756 HSA "
a, 58 1891 pEE12:HSA.sTR6 Soluble mature TR6 fused downstream of pEE12.1 274 58 490 757 758 HSA
H
s CA
w HSA.
(5) Lo 59 1892 pEE12:sTR6.HSA Synthetic full length TR6 fused upstream pEE12.1 275 59 491 759 760 TR6 I.) of mature HSA.

a, 60 1906 pC4:PTH.S1-F34.HSA Amino acids Si to F34 of PTH fused pC4 276 60 492 761 762 MPIF 0 (5) ' (junctioned) upstream of mature HSA and downstream I.) of MPIF leader sequence. There are two H
cloning junction amino acids (T,S) between PTH and HSA.
61 1907 pC4:HSA.PTH.S1-F34 Amino acids 51 to F34 fused downstream pC4 277 61 493 763 764 HSA
(junctioned) of FL HSA. The last C-terminal amino acid (L) residue is missing for HSA in the cloning junction between HSA and PTH.
Iv 62 1912 pC4:sTR6.HSA Synthetic full length TR6 fused upstream pC4 278 62 494 765 766 Native TR6 n ,-i of mature HSA.
leader -----63 1913 pC4:HSA.synTR6.V30- Amino acids V30 to H300 of synthetic pC4 279 63 495 767 768 HSA cp o t..) H300 (seamless) TR6 (shown as Vito H271 of SEQ ID
.1-.
o NO:495) fused downstream of full-length oc, o HSA.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ .SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B
X
A
64 1914 pC4:PTH.S1-F34.HSA Amino acids Si to F34 of PTH fused pC4 280 64 496 769 770 MPIF
(seamless) downstream of MPIF leader sequence and upstream of mature HSA.
65 1916 pC4:HSA.KGF2D28.A6 Amino acids A63 to S208 of full length pC4 281 65 497 771 772 HSA
3-S208 KGF2 fused downstream of HSA.
66 1917 pC4:KGF2D28.A63- Amino acids A63 to S208 of KGF2 fused pC4 282 66 498 773 774 HSA/kex2 S208:HSA upstream of mature HSA.
67 1925 pcDNA3.EPO Ml- Amino acids M1 to D192 of EPO variant pcDNA3 283 67 499 775 776 Native 0 D192.HSA (where glycine at amino acid 140 has been EPO leader (J.) replaced with an arginine) fused upstream peptide (5) of HSA. D192 of EPO and D1 of mature HSA are the same amino acids in this construct.

68 1926 pcDNA3:SPHSA.EPO Amino acids A28 to D192 of EPO variant pcDNA3 284 68 500 777 778 MPIF (5) A28-D192 (where glycine at amino acid 140 has been replaced with an arginine) fused upstream of mature HSA and downstream of the MPIF leader peptide.
69 1932 pEE12.1:HSA.PTH.S1- Amino acids 1 to 34 of PTH fused pEE12.1 285 69 501 779 780 HSA
F34 downstream of full length HSA.
70 1933 pSAC35:HCC-1.T20- Amino acids T20 to N93 of HCC-1 fused pSAC35 286 70 502 781 782 HSA/kex2 N93:HSA upstream of mature HSA and downstream 1-d of the HSA/kex2 leader sequence. -oe Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader c.....,_4 o No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o 1¨

X
A
71 1934 pSAC35:HCC- Amino acids T20 to N93 of HCC-1 fused pSAC35 287 71 503 783 784 HSA/kex2 1C.O.T20-N93:HSA upstream of mature HSA and downstream of the HSA/kex2 leader sequence. DNA
sequence is codon optimized for yeast expression.
72 1938 pEE12.1:PTH.S1- Amino acids S1 to F34 of PTH fused pEE12.1 288 72 504 785 786 MPIF
n F34.HSA upstream of mature HSA and downstream of MPIF leader sequence.

I.) .1, 73 1941 pC4:HSA/PTH84 PTH fused downstream of full length pC4 289 73 505 787 788 HSA
H
---ACA
-{. (junctioned) HSA. The last amino acid of HSA (Leu) (5) co has been deleted.
I.) 74 1947 pSAC35:d8HCC-1.G28- Amino acids G28 to N93 of HCC-1 fused pSAC35 290 74 506 789 790 HSA/kex2 0 .1, N93:HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence.
(5) I.) 75 1948 pSAC35:d8HCC- Amino acids G28 to N93 of HCC4 fused pSAC35 291 75 507 791 792 HSA/kex2 H
1 C. 0 . G28-N93:HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence. DNA
_ sequence is codon optimized for yeast expression.
76 1949 pC4:PTH.S1-Q84/HSA PTH fused downstream of the MPIF pC4 (junctioned) leader sequence and upstream of mature HSA. There are two additional amino 1-d n acids between PTH84 and HSA as a result of the cloning site.
cp o 77 1952 pcDNA3.11L2.HSA Full length human IL-2, having a Cysteine pCDNA3.1 293 77 509 795 796 Native IL-2 !=1-.,..,' to Serine mutation at amino acid 145, leader =
oe o fused upstream of mature HSA.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence S
NO:Y NO: NO:Z NO: NO:B
X
A
78 1954 pC4:1L2.HSA Full length human IL-2, having a Cysteine pC4 294 78 510 797 798 Native IL-2 to Serine mutation at amino acid 145, leader fused upstream of mature HSA.
79 1955 pSAC35:t9HCC-1.G28- Amino acids 028 to N93 of HCC-1 fused pSAC35 295 79 511 799 800 HSA/kex2 N93:spcHSA upstream of a 16 amino acid spacer and mature HSA and downstream of HSA/kex2 leader sequence.

80 1956 pSAC35:HSA.scFv116 Single chain BLyS antibody fused pSAC35 296 80 512 801 802 HSA/kex2 A01 downstream of HSA with HSA/kex2 leader sequence. This construct also (5) contains a His tag at the 3' end.

81 1966 pC4:EPO.M1- Amino acids M1 to D192 of EPO variant pC4 297 81 513 Native 0 D192.HSA (where glycine at amino acid 140 has been EPO leader 0 (5) replaced with an arginine) fused upstream peptide Construct is also named of mature HSA.
pC4:EPOM1-D192.HSA
82 1969 pC4:MP1Fsp.HSA.EPO. Amino acids A28 to D192 of EPO variant pC4 298 82 514 MPIE
A28-D192 (where glycine at amino acid 140 has been replaced with an arginine) fused downstream of MPIF leader sequence and 1-d upstream of mature HSA.
83 1980 pC4:EPO.A28-D192. Amino acids A28 to D192 of EPO variant pC4 299 83 515 803 804 HSA
HSA (where glycine at amino acid 140 has been replaced with an arginine) fused downstream of the HSA leader peptide oe and upstream of mature HSA.

Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader st...) No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
84 1981 pC4.HSA-EPO.A28- Amino acids A28 to D192 of EPO variant pC4 300 84 516 805 806 HSA
D192. (where glycine at amino acid 140 has been replaced with an arginine) fused downstream of the full length HSA.
85 1989 pSAC35:activeAC2inhi Active inhibitor of ACE2 (DX512) fused pSAC35 301 85 517 807 808 HSA/kex2 bitor:HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence.

86 1994 pEE12.1.HSA- Amino acids A28 to D192 of EPO variant pEE12.1 302 86 518 HSA
EPO.A28-D192. (where glycine at amino acid 140 has been Fa replaced with an arginine) fused (5) downstream of full length HSA.

D192. HSA (where glycine at amino acid 140 has been replaced with an arginine) fused (5) downstream of the HSA leader peptide and upstream of mature HSA.
88 1996 pEE12.1:MPIFsp.HSA. Amino acids A28 to D192 of EPO variant pEE12.1 304 88 520 MPIF
EPO.A28-D192 (where glycine at amino acid 140 has been replaced with an arginine) fused downstream of MPIF leader sequence and upstream of mature HSA.
D192.HSA (where glycine at amino acid 140 has been EPO leader Lt replaced with an arginine) fused upstream of mature HSA.
oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader F,3 No. ID VectorO' ID ID ID ID ID Sequence o o NO:Y NO: NO:Z NO: NO:B
=

X
A 1.., 90 1998 pC4:CKB1.G28- Amino acids G28 to N93 of CkBetal pC4 306 90 522 809 810 HSA
N93.HSA fused upstream of mature HSA and downstream of the HSA leader sequence.
91 2000 pSAC35:HSA:activeAC Active inhibitor of ACE2 (DX512) fused pSAC35 307 91 523 811 812 HSA
2inhibitor downstream of HSA.
92 2001 pSAC35:inactiveAC2in Inactive inhibitor of ACE2 (DX510) pSAC35 308 92 524 813 814 HSAikex2 n hibitor:HSA fused upstream of mature HSA and downstream of HSA/kex2 leader I.) a, sequence.
, H
93 2002 pSAC35:HSA.inactiveA Inactive inhibitor of ACE2 (DX510) pSAC35 309 93 525 815 816 HSA Lo (5) C2inhibitor fused downstream of HSA.
Lo I.) 94 2011 pC4:IFNb-HSA Full length IFNb fused upstream of pC4 310 94 526 817 818 Native 0 mature HSA.
IFNb a, leader (5) 95 2013 pC4:HSA-IFNb.M22- Amino acids M22 to N187 of IFNb pC4 311 95 527 HSA I.) H
N187 (fragment shown as amino acids M1 to N166 of SEQ ID NO:527) fused downstream of HSA.
96 2016 pC4:TR1.M1- Amino acids M1 to L401 of TR1 fused pC4 312 96 528 819 820 Native TR1 L401.HSA upstream of mature HSA. Native TR1 signal sequence used. A Kozak sequence was added.
1-d n 97 2017 pC4:HSA.TR1.E22- Amino acids E22 to L401 of TR1 fused pC4 313 97 529 821 822 HSA
L401 downstream of HSA.
cp =
98 2021 pC4:PTH.S1-Q84/HSA Amino acids 1-84 of PTH fused upstream pC4 314 98 530 823 824 HSA t..) .1-.
(seamless) of mature HSA and downstream of native =
oe vD
HSA leader sequence.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID
Vector-a-, ID ID ID ID ID Sequence o o NO:Y NO: NO:Z NO: NO:B
=

X
, A 1.., 99 2022 pEE12.1:PTH.S1- Amino acids 1-84 of PTH fused upstream pEE12.1 315 99 531 HSA
Q84.HSA of mature HSA and downstream of native HSA leader sequence.
100 2023 pSAC35.PTH.S1- Amino acids 1-84 of PTH fused upstream pSAC35 316 100 532 825 826 HSA/kex2 Q84.HSA of mature HSA and downstream of HSA/kex2 leader sequence.
101 2025 pSAC35:teprotide.space Teprotide fused upstream of a linker and pSAC35 317 101 533 827 828 n r.HSA mature HSA.

I.) 102 2026 pSAC35:HSA.spacer.tep Teprotide fused downstream of HSA
and pSAC35 318 102 534 829 830 HSA a, -,1 H
'---) 00 rotide a linker.
u.) (5) 103 2030 pSAC35.ycolL-2.A21- Amino acids A21 to T153 of 1L-2 fused pSAC35 319 103 535 831 832 HSA/kex2 u.) I.) T153.HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence. DNA
a, encoding IL-2 has been codon optimized.
(5) 104 2031 pSAC35.HSA.ycolL- Amino acids A21 to T153 of IL-2 fused pSAC35 320 104 536 833 834 HSA/kex2 I.) H
2.A21-T153 downstream of HSA with the HSA/kex2 leader sequence. DNA encoding 1L-2 has been codon optimized.
105 2047 pC4HSA:SP.EPO A28- Amino acids A28 to D192 of EPO variant pSAC35 321 105 537 835 836 MPIF
D192.HSA (where glycine at amino acid 140 has been replaced with an arginine) fused upstream of mature HSA and downstream of MPIF
1-d n leader peptide.
106 2053 pEE12:IFNb-HSA Full length IFNb fused upstream of pEE12.1 322 106 538 Native cp =
mature HSA.
IFNb t..) also named named leader =
oe pEE12.1:IFN[3-HSA
vD


Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader st,' o No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B o 1.., X
A
107 2054 pEE12:HSA-lFNb Mature IFNb fused downstream of HSA.
pEE12.1 323 107 539 HSA
108 2066 pC4:GM-CSF.M1- Amino acids M1 to E144 of GM-CSF pC4 324 108 540 837 838 Native E144.HSA fused upstream of mature HSA.
GM-CSF
109 2067 pC4:HSA.GM- Amino acids A18 to E144 of GM-CSF
pC4 325 109 541 839 840 HSA
CSF.A18-E144 fused downstream of HSA.
110 2085 pEE12.1:TR1.M1- Amino acids M1 to L401 of TR1 fused pEE12.1 326 110 542 Native TR- n L401.HSA upstream of mature HSA.

111 2086 pEE12.1:HSA.TR1.E22- Amino acids E22 to L401 (fragment pEE12.1 327 111 543 HSA "
a, L401 shown as amino acids El to L380 of SEQ
H

CA
ID NO:543) of TR1 fused downstream of (5) co HSA.
I.) 112 2095 pC4:HSA-BLyS.A134 Amino acids A134 to L285 of BLyS fused pC4 328 112 544 841 842 HSA 0 a, downstream of HSA.

(5) 113 2096 pC4:sp.BLyS.A134- Amino acids A134 to L285 of BLyS
pC4 329 113 545 843 844 Native I.) L285.HSA (fragment shown as amino acids Al to L152 of SEQ ID NO:545) fused upstream of mature HSA and downstream of the CKb8 signal peptide.
114 2101 pcDNA3:SP.Ck7 Q22- N-terminal Methionine fused to amino pcDNA3 330 114 546 845 846 MPIF
A89.HSA. acids Q22 to A89 of Ck(37 fused upstream of mature HSA and downstream of MPIF
1-d signal peptide.
n 1-i 115 2102 pEE12.1:SP.EPO A28- Amino acids A28 to D192 of EPO variant pEE12.1 331 115 547 MPIF
c) D192.HSA (where glycine at amino acid 140 has been o t..) replaced with an arginine) fused upstream .1-.
o of mature HSA and downstream of MPIF
ce o leader peptide.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence E
NO:Y NO: NO:Z NO: NO:B
X
A , 116 2129 pC4:TR2.M1- Amino acids M1 -A192 of TR2 fused pC4 332 116 548 847 848 Native TR2 A192.HSA upstream of HSA.
117 2137 pSAC35.MDC.G25- Amino acids G25 to Q93 of MDC fused pSAC35 333 117 549 849 850 HSA/kex2 Q93.HSA. upstream of mature HSA and downstream of HSA/kex2 leader sequence.
118 2141 HSA-CK-Beta4 Full length CK-beta4 fused downstream pSAC35 334 118 550 851 852 HSA
of HSA.

119 2146 pC4:Leptin.HSA Full length Leptin fused upstream of pC4 335 119 551 853 854 Native mature HSA.
leptin 120 2181 pC4:HSAIL1Ra(R8- Amino acids R8 to E159 of IL1Ra (plus pC4 336 120 552 855 856 HSA
(5) E159) an added methionine at N-terminus) fused downstream of HSA.

121 2182 pC4:MPIFsp(M1- Amino acids R8 to E159 of IL1Ra (plus pC4 337 121 553 857 858 MPIF 0 (5) A21),IL1Ra(R8- an added methionine at N-terminus) fused E159).HSA downstream of the MPIF leader sequence and upstream of mature HSA.
122 2183 pSAC35:HSAIL1Ra(R8 Amino acids R8 to E159 of IL1Ra (plus pSAC35 338 122 554 859 860 HSA
-E159) an added methionine at N-terminus) fused downstream of HSA.
123 2184 pC4:HSA.Leptin.V22- Amino acids V22 to C167 of Leptin fused pC4 339 123 555 861 862 HSA
C166 downstream of HSA.
1-d 124 2185 pSAC35:Th1Ra(R8- Amino acids R8 to E159 of IL1Ra (plus pSAC35 340 124 556 863 864 HSA/kex2 El 59).HSA an added methionine at N-terminus) fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B
X
A
125 2186 pSAC35:Leptin.V22- Amino acids V22 to C167 of Leptin fused pSAC35 341 125 557 865 866 HSA/kex2 C166.HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence.
126 2187 pSAC35:HSA.Leptin.V Amino acids V22 to C167 of Leptin pSAC35 342 126 558 867 868 HSA/kex2 22-C166 fused downstream of HSA with HSA/kex2 leader sequence.
127 2226 pcDNA3(+):TREM- Amino acids A21 to P202 of TREM-1 pCDNA3.1 343 127 559 869 870 MPIF
1(21-202)-HSA fused upstream of mature HSA and downstream of the MPIF leader sequence.
,19 128 2230 pC4:TREM-1.M1- Amino acids M1 to P202 of TREM-1 pC4 344 128 560 871 872 Native (5) P202.HSA fused upstream of mature HSA.

129 2240 pC4:SP.Ck7 Q22- N-terminal Methionine fused to amino pC4 345 129 561 873 874 MPIF 0 A89.HSA. acids Q22 to A89 of Ckf37 fused upstream of mature HSA and downstream of the (5) MPIF leader sequence. Contains a linker sequence between Ck137 and HSA.
130 2241 pC4:HSA.Ck7metQ22- N-terminal Methionine fused to amino pC4 346 130 562 875 876 HSA/kex2 A89. acids Q22 to A89 of Chemokine beta 7 (Ckbeta 7 or CK7) fused downstream of HSA with HSA/kex2 leader sequence.
Contains a linker sequence between CkB7 and HSA.
131 2244 pC4.HCNCA73.HSA HCNCA73 fused upstream of mature pC4 HSA.
oe Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: = NO:B
X
A
132 2245 pScNHSA:CK7.Q22- Amino acids Q22 to A89 of Ck137 fused pScNHSA 348 132 564 879 880 HSA/kex2 A89 downstream of HSA with HSA/kex2 leader sequence. Contains a linker sequence between Clc137 and HSA.
133 2246 pScCHSA.CK7metQ22- N-terminal Methionine fused to amino pScCHSA 349 133 565 881 882 HSA/kex2 A89 acids Q22 to A89 of Ckf37 fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.

134 2247 pSAC35:CK7metQ22- N-terminal Methionine fused to amino pSAC35 350 134 566 883 884 HSA/kex2 00 A89.HSA. acids Q22 to A89 of C1437 fused upstream (5) of mature HSA and downstream of HSA/kex2 leader sequence.

135 2248 pSAC35:HSA.CK7met N-terminal Methionine fused to amino pSAC35 351 135 567 885 886 HSA/kex2 Q22-A89. = acids Q22 to A89 of Ck137 fused (5) downstream of HSA with HSA/kex2 leader sequence. Contains a linker sequence between Ck137 and HSA.
136 2249 pSAC35:IFNa2-HSA Mature 1FNa2 fused upstream of mature pSAC35 352 136 568 887 888 HSA/kex2 HSA and downstream of HSA/kex2 leader also named: sequence.
pSAC23 :1FNa2-HSA
137 2250 pSAC35:HSA.INSUL1N Mature Insulin wherein the C-peptide is pSAC35 353 137 569 889 890 HSA 1-d (GYG) replaced by the C-domain of IGF-1 fused downstream of HSA. DNA encoding also named: Insulin was codon optimized.
pSAC35.HSA.I4SULIN
c'e G(GYG).F1-N62 Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence g;
NO:Y NO: NO:Z NO: NO:B
X
A
138 2251 pScCHSA:VEGF2.T103 Amino acids T103 to R227 of VEGF2 pScCHSA 354 138 570 891 892 HSA/kex2 -R227. fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
139 2252 pScNHSA:VEGF2.T103 Amino acids T103 to R227 of VEGF2 pScNHSA 355 139 571 893 894 HSAJkex2 -R227. fused downstream of HSA with HSA/kex2 leader sequence.

140 2255 pSAC35:11\TSULIN(G Mature Insulin wherein the C-peptide is pSAC35 356 140 572 895 896 HSA/kex2 YG).HSA replaced by the C-domain of IGF-1 fused tka upstream of mature HSA and downstream (5) also named of HSA/kex2 leader. DNA encoding pSAC35.INSULING(G Insulin was codon optimized.

YG).F1-N62.HSA

(5) 141 2256 pSAC35:VEGF2.T103- Amino acids T103 to R227 of VEGF2 pSAC35 357 141 573 897 898 HSA/kex2 R227.HSA fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
142 2257 pSAC35:HSA.VEGF2.T Amino acids T103 to R227 of VEGF-2 pSAC35 358 142 574 899 900 HSA/kex2 103-R227 fused downstream of HSA with HSA/kex2 leader sequence.
143 2271 pEE12.1:HCHNF25M1- Amino acids M1 to R104 of HCHNF25 pEE12.1 359 143 575 Native 1-d R104.HSA fused upstream of mature HSA.

c) oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
144 2276 pSAC35:HSAINSULIN Mature Insulin wherein the C-peptide is pSAC35 360 144 576 901 902 HSA
(GGG) replaced by a synthetic linker fused downstream of HSA. DNA encoding also named: Insulin was codon optimized.
pSAC35.HSA.INSULIN
G(GGG).F1-N58 145 2278 pSAC35:insulin(GGG). Mature Insulin wherein the C-peptide is pSAC35 361 145 577 903 904 HSA/kex2 HSA replaced by a synthetic linker fused downstream of HSA/kex2 leader and 00 upstream of mature HSA. DNA encoding (5) Insulin was codon optimized.
146 2280 pC4:HCHNF25.HSA HCHNF25 fused upstream of mature pC4 362 146 578 905 906 Native 0 HSA.

147 2283 pScCHSA:EPOcoA28- Amino acids A28 to D192 of EPO variant pScCHSA 363 147 579 907 908 HSA/kex2 D192.51N/Q, 65N/Q, (where glycine at amino acid 140 has been 110N/Q EPO replaced with an arginine) are fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
Glycosylation sites at amino acids 51, 65 and 110 are mutated from N to Q residue.
DNA encoding EPO is codon optimized.
1-d oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader (..) No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
148 2284 pScNHSA:EPOcoA28- Amino acids A28 to D192 of EPO variant pScNHSA 364 148 580 909 910 HSA/kex2 D192.51N/Q, 65N/Q, (where glycine at amino acid 140 has been 110N/Q EPO replaced with an arginine) fused downstream of mature HSA and HSA/kex2 leader sequence. Glycosylation sites at amino acids 51, 65 and 110 are mutated from N to Q residue. DNA
encoding EPO is codon optimized.

149 2287 pSAC35:EPOc0A28- Amino acids A28 to D192 of EPO variant pSAC35 365 149 581 911 912 HSA/kex2 00 D192.51N/Q,65N/Q,110 (where glycine at amino acid 140 has been (5) N/Q.HSA. replaced with an arginine) fused upstream of mature HSA and downstream of HSA/kex2 leader sequence. Glycosylation sites at amino acid 51, 65 and 110 are (5) mutated from N to Q residue. DNA
encoding EPO is codon optimized.
150 2289 pSAC35:HSA.EPOcoA Amino acids A28 to D192 of EPO variant pSAC35 366 150 582 913 914 HSA/kex2 28- (where glycine at amino acid 140 has been D192.51N/Q,65N/Q,110 replaced with an arginine) fused.
N/Q. downstream of mature HSA and HSA/kex2 leader sequence. Glycosylation sites at amino acid 51, 65 and 110 are 1-d mutated from N to Q residue. DNA
encoding EPO is codon optimized.
oe Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader st...) No. ID Vector ID ID ID ID ID Sequence E
NO:Y NO: NO:Z NO: NO:B
X
A
151 2294 pC4:EPO.R140G.HSA Amino acids M1-D192 of EPO fused pC4 367 151 587 915 916 Native upstream of mature HSA. The EPO
EPO
also named sequence included in construct 1997 was pC4.EPO.R1406.HSA used to generate this construct, mutating arginine at EPO amino acid 140 to glycine. This mutated sequence matches the wildtype EPO sequence.
152 2295 pSAC35:humanresistin. Amino acids 1(19 to P108 of Resistin pSAC35 368 152 584 917 918 HSA/kex2 0 K19-P108:HSA fused upstream of mature HSA and oo downstream of HSA/kex2 leader (5) sequence.
153 2296 pSAC35:HSA:humanres Amino acids K19 to P108 of Resistin pSAC35 369 153 585 919 920 HSA 0 istin.K19-P108 fused downstream of HSA.

154 2297 pSAC35:humanresistin. Amino acids K19 to P108 of Resistin pSAC35 370 154 586 921 922 HSA/kex2 (5) K19-P108.stop:HSA fused upstream of mature HSA and downstream of HSA/kex2 leader sequence. Includes two stops at 3' end for termination of translation before the HSA.
155 2298 pEE12.1:EPO.R140G.H Amino acids M1 to D192 of EPO fused pEE12.1 371 155 587 923 924 Native SA upstream of mature HSA. The EPO
EPO
sequence included in construct 1997 was used to generate this construct, mutating arginine at EPO amino acid 140 to glycine. This mutated sequence matches the wildtype EPO sequence.
oe . .
Table 2 = 0 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader F...4 No. ID Vector ID ID ID ID ID Sequence E
NO:Y NO: NO:Z NO: NO:B o --.1 1¨

X
A
156 2300 pC4:humanresistin.M1- Amino acids M1 to P108 of Resistin fused pC4 372 156 588 925 926 Native P108:HSA upstream of mature HSA.
resistin 157 2309 pEE12.1:humanresistin. Amino acids M1 to P108 of Resistin fused pEE12.1 373 157 589 927 Native M1-P108:HSA upstream of mature HSA.
resistin 158 2310 pc4:EPOco.M1- Amino acids M1 to D192 of EPO variant pC4 374 158 590 928 929 Native D192.HSA fused upstream of mature HSA. DNA
EPO n encoding EPO is codon optimized. The EPO sequence included in construct 1997 I.) a, was used to generate this construct, H

--.1 mutating arginine at EPO amino acid 140 (5) u.) to glycine. This mutated sequence I.) matches the wildtype EPO sequence.

a, 159 2311 pC4:EPO.M1- Amino acids M1 to D192 of EPO fused pC4 375 159 591 930 931 Native 1 G27.EPOco.A28- upstream of mature HSA. DNA encoding EPO (5) I.) D192.HSA only EPO portion is codon optimized.
H
The EPO sequence included in construct 1997 was used to generate this construct, mutating arginine at EPO amino acid 140 to glycine. This mutated sequence matches the vvildtype EPO sequence.
160 2320 pC4:HCHNF25M1- Amino acids M1 to R104 of HCHNF25 pC4 376 160 592 932 933 Native R104.HSA fused upstream of mature HSA.
HCHNF25 e n 161 2325 pC4.EPO:M1- Amino acids M1 to D192 of EPO fused pC4 377 161 593 Native D192.HSA.Codon opt. upstream of mature HSA. DNA encoding EPO cp o EPO is codon optimized.
t..) =
oe o 1¨

, =
Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader F...4 o No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o 1¨

X
A
162 2326 pEE12.1.EPO:M1- Amino acids M1 to D192 of EPO fused pEE12.1 378 162 594 Native D192.HSA.Codon opt. upstream of mature HSA. DNA encoding EPO
EPO is codon optimized.
, 163 2328 pC4:HLDOU18.K23- Amino acids 1(23 to R429 of HLDOU18 pC4 379 163 595 934 935 HSA
R429.HSA fused upstream of mature HSA and downstream of native HSA leader sequence.
164 2330 CK-Beta4-HSA Full length Ckbeta4 fused upstream of pSAC35 380 164 596 936 937 Native 0 I.) a, mature HSA.

H
gg 165 2335 pC4:MP1Fsp.ck{b}4D3 Amino acids D31 to M96 of Ckbeta4 pC4 381 165 597 938 939 MPIF co (5) co 1-M96.HSA fused upstream of mature HSA and I.) downstream of MPIF leader sequence.

a, 166 2336 pC4:MPlFsp.ck{b}4G3 Amino acids G35 to M96 of Ckbeta4 pC4 382 166 598 940 941 MPIF 0 5-M96.HSA fused upstream of mature HSA and (5) I.) downstream of MPIF leader sequence.
H
167 2337 pC4:MPIFsp.ck{b}4G4 Amino acids G48 to M96 of Ckbeta4 pC4 383 167 599 942 943 MPIF
8-M96.HSA fused upstream of mature HSA and downstream of MPIF leader sequence.
168 2338 pC4:MPIFsp.ck{b}4A6 Amino acids A62 to M96 of Ckbeta4 pC4 384 168 600 944 945 MPIF
2-M96.HSA fused upstream of mature HSA and downstream of MPIF leader sequence.
169 2340 pC4:HSA.HLDOU18.K Amino acids 1(23 to R429 of HLDOU18 pC4 385 169 601 946 947 HSA 1-d n 23-R429 fused downstream of HSA.
170 2343 pSAC35.INV- Mature Interferon alpha2 fused upstream pSAC35 386 170 602 948 949 invertase ci) o IFNA2.HSA of mature HSA and downstream of t..) invertase signal peptide.
o oo o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B

X
A 1.., 171 2344 pC4.SpIg.EPO:A28- Amino acids A28 to D192 of EPO fused pC4 387 171 603 950 951 Mouse Ig D192.HSA.Codon opt. upstream of mature HSA and downstream leader of mouse Ig leader sequence. DNA
encoding EPO is codon optimized.
172 2348 pC4:MPIFsp.ck{b}4G5 Amino acids G57 to M96 of Ckbeta4 pC4 388 172 604 952 953 MPIF
7-M96.HSA fused upstream of mature HSA and n downstream of MPIF leader sequence.
173 2350 pC4:MPIFsp.HLDOU18 Amino acids S320 to R429 of HLDOU18 pC4 I.) (S320-R429).HSA fused upstream of mature HSA and a, -,1 H

..o downstream of MPIF leader sequence.
Lo (5) 174 2351 pC4:HSA.HLDOU18(S Amino acids S320 to R429 of HLDOU18 pC4 390 174 606 956 957 HSA Lo I.) 320-R429) fused downstream of HSA.

175 2355 pSAC35:MATalpha.d8c Amino acids G28 to N93 of Ckbetal pSAC35 391 175 607 958 959 MFa-1 a, kbetal .G28-N93:HSA fused upstream of mature HSA and (5) downstream of the yeast mating factor I.) H
alpha leader sequence.
176 2359 pEE12:HLDOU18.K23- Amino acids 1(23 to R429 of HLDOU18 pEE12.1 392 176 608 HSA
R429.HSA fused upstream of mature HSA and downstream of native HSA leader sequence.
177 2361 pC4:HRDFD27:HSA HRDFD27 fused upstream of mature pC4 393 177 609 960 961 Native HSA.
HRDFD27 't n 178 2362 pEE12:HSA.HLDOU18. Amino acids 1(23 to R429 of HLDOU18 pEE12.1 394 178 610 HSA
K23-R429 fused downstream of HSA.
cp o t..) =
oe o 1¨

Table 2 =

_______________________________________________________________________________ _________________________________________ o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence g;
o NO:Y NO: NO:Z NO: NO:B

1.., X
A
179 2363 pC4GCSF.HSA.EPO.A Amino acids M1 to P204 of GCSF fused pC4 395 179 611 Native 28-D192 upstream of mature HSA which is fused GCSF
upstream of amino acids A28 to D192 of EPO variant (where amino acid 140 of EPO is mutated from glycine to arginine.) 180 2365 pEE12.1.HCNCA73HS HCNCA73 is fused upstream of mature pEE12.1 396 180 612 962 963 Native n A HSA.

181 2366 pSAC35.MAF- Mature lFNa2 fused upstream of mature PSAC35 397 181 613 964 965 MF'a-1 N) a, IFNa2.HSA HSA and downstream of yeast mating H
µ.0CA
c) factor alpha leader sequence.
(5) co 182 2367 pEE12.MPIFsp.HLDOU Amino acids S320 to R429 of HLDOU18 pEE12.1 398 182 614 966 967 MPIF I.) 18.S320-R429.HSA fused upstream of mature HSA and a, downstream of MPIF leader sequence.

(5) 183 2369 pC4:HLDOU18.HSA Amino acids M1 to R429 of HLDOU18 pC4 399 183 615 968 969 Native I.) fused upstream of mature HSA.

184 2370 pEE12:HLDOU18.HSA Amino acids M1 to R429 of HLDOU18 pEE12.1 400 184 616 Native fused upstream of mature HSA.

185 2373 pC4.GCSF.HSA.EPO.A Amino acids M1 to P204 of GCSF is pC4 401 185 617 Native 28-D192.R1400 fused upstream of mature HSA which is GCSF
fused upstream of amino acids A28 to D192 of EPO, wherein amino acid 140 is 1-d glycine. The EPO sequence included in n 1-i construct 1997 was used to generate this c) construct, mutating arginine at EPO amino o acid 140 to glycine. This mutated t..) o sequence matches the wildtype EPO
oe o sequence.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader (..) No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
186 2381 pC4:HSA-IFNa2(C17- Amino acids C17 to E181 of IFNa2 pC4 El 81) (fragment shown as amino acids Cl to E165 of SEQ ID NO:618) fused downstream of HSA.
187 2382 pC4:IFNa2-HSA IFNa2 fused upstream of mature HSA.
pC4 403 187 619 972 973 Native leader 188 2387 pC4:EPO(G140)-HSA- Amino acids M1 -D192 of-EPO fused pC4 404 188 620 Native 0 GCSF.T31-P204 upstream of mature HSA which is fused EPO
upstream of amino acids T31 to P204 of (5) GCSF.
189 2407 pC4:HWHGZ51.M1- Amino acids M1 to N323 of HWHGZ51 pC4 405 189 621 974 975 Native 0 N323.HSA fused upstream of mature HSA.

(5) 190 2408 pEE12.1:HWHGZ51.M Amino acids M1 to N323 of HWHGZ51 pEE12.1 406 190 622 976 977 Native 1-N323.HSA fused upstream of mature HSA.

191 2410 pSAC351NV:IFNa-HSA Mature IFNa2 fused downstream of the pSAC35 407 191 623 978 979 invertase invertase signal peptide and upstream of mature HSA.
192 2412 pSAC35:de1KEX.d8ckb Amino acids G28 to N93 of Ckbetal pSAC35 408 192 624 980 981 HSA minus etal.G28-N93:HSA fused downstream of the HSA signal the KEX
sequence (with the KEX site deleted ¨ last site 6 amino acids of the leader) and upstream of mature HSA.
oe Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence S
o NO:Y NO: NO:Z NO: NO:B
--.1 1¨

X
A
193 2414 pC4.EPO:M1- Amino acids M1 to D192 of EPO fused pC4 409 193 625 982 983 Native D192copt.HSA.GCSF.T upstream of mature HSA which is fused EPO
31-P204 upstream of amino acids T31 to P204 of GCSF. DNA encoding EPO has been also named: codon optimized.
pC4.EPO:M1-n D192copt.HAS.GCSF.T

I.) 194 2428 pN4:PTH.S1-Q84/HSA Amino acids Si to Q84 of PTH fused pN4 410 194 626 HSA
H
upstream of mature HSA and downstream co (5) IQ co of the native HSA leader sequence.
_ I.) 195 2441 pEE12.EPO:M1- Amino acids M1 to D192 of EPO fused pEE12.1 409 196 628 EPO leader 0 .1,.

D192copt.HSA.GCSF.T upstream of mature HSA which is fused 31-P204 upstream of amino acids T31 to P204 of (5) I.) GCSF. DNA encoding EPO has been H
also named: codon optimized.
pEE12.EPO:M1-D192copt.HAS.GCSF.T

196 2447 pC4:HSA.humancalcito Amino acids C98 to G130 of SEQ ID
pC4 413 197 629 986 987 HSA
nin.C1-G33 NO:629 fused downstream of HSA.
197 2448 pSAC35:GLP-1(7- Amino acids H98 to R127 of pSAC35 414 198 630 988 989 HSA/kex2 A
36).HSA preproglucagon (SEQ ID NO:630) (hereinafter this specific domain will be cp o referred to as "GLP-1(7-36)") is fused t..) .1-.
upstream of mature HSA and downstream c, oe o of HSA/kex2 leader sequence.


Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence S
o NO:Y NO: NO:Z NO: NO:B

1.., X
A
198 2449 pSAC35:INV.d8CKB1. Amino acids G28 to N93 of Ckbetal pSAC35 415 199 631 990 991 Invertase G28-N93 :HSA fused downstream of the invertase signal peptide and upstream of mature HSA.
199 2455 pSAC35:HSA.GLP-1(7- GLP-1(7-36) is fused downstream of pSAC35 416 200 632 992 993 HSA/kex2 36) mature HSA and HSA/kex2 leader sequence.
n 200 2456 pSAC35:GLP-1(7- Amino acids 1198 to R127 of pSAC35 417 201 633 994 995 HSA/kex2 36(A8G)).HSA Preproglucagon (SEQ ID NO:633)(also "
a, referred to as "GLP-1(7-36)") is mutated H
v:) at a-mino acid 99 of SEQ ID NO:633 to u.) (5) t.,.) u.) replace the alanine with a glycine. This I.) particular GLP-1 mutant will be a, ' hereinafter referred to as "GLP-1(7-36(A8G))" and corresponds to the (5) I.) sequence shown in SEQ ID NO:1808.
H
GLP-1(7-36(A8G)) is fused upstream of mature HSA and downstream of HSA/kex2 leader sequence.
201 2457 pSAC35:HSA.GLP-1(7- GLP-1(7-36(A8G)) (SEQ ID NO:1808) is pSAC35 418 202 634 996 997 HSA/kex2 36(A8G)) fused downstream of mature HSA and HSA/kex2 leader sequence.
202 2469 pSAC35:HSA.exendin. Amino acids 1148 to S86 of Extendin pSAC35 419 203 635 HSA Iv n H48-S86 fused downstream of full length HSA.
203 2470 pSAC35:Exendin.H48- Amino acids 1148 to S86 of Extendin pSAC35 420 204 636 HSA/kex2 ci) o S86.HSA fused upstream of mature HSA and t..) .1-.
downstream of HSA/kex2 leader ' oe o sequence.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
204 2473 pC4.1-11,DOU18:HSA:S Ml-R319 of HLDOU18 (containing the pC4 421 205 637 998 999 Native 320-R429 furin site RRKR) followed by residues 'LE' followed by mature HSA followed by 'LE and amino acids S320 through R429 of HLDOU18 (fragment shown as SEQ ID NO:637).
205 2474 pSAC35.MDC.P26- Amino acids P26 to Q93 of MDC fused pSAC35 422 206 638 1000 1001 HSA/kex2 Q93.HSA downstream of the HSA/kex2 leader and upstream of mature HSA.
.4.') 206 2475 pSAC35.MDC.M26-Amino acids Y27 to Q93 of MDC with an pSAC35 423 207 639 1002 1003 HSA/kex2 (5) Q93.HSA N-terminal methionine, fused downstream of the HSA/kex2 leader and upstream of mature HSA.

207 2476 pSAC35.MDC.Y27- Amino acids Y27 to Q93 of MDC fused pSAC35 424 208 640 1004 1005 HSA/kex2 (5) Q93.HSA downstream of the HSA/kex2 leader and upstream of mature HSA.
208 2477 pSAC35.MDC.M27- Amino acids G28 to Q93 of MDC with an pSAC35 425 209 641 1006 1007 HSA/kex2 Q93.HSA N-terminal methionine, fused downstream of the HSA/kex2 leader and upstream of mature HSA.
209 2489 pSAC35:HSA.C17.A20- Amino acids A20 to R136 of C17 fused pSAC35 426 210 642 1008 1009 HSA/kex2 R136 downstream of mature HSA with 1-d HSA/kex2 leader sequence.
210 2490 pSAC35:C17.A20- Amino acids A20 to R136 of C17 fused pSAC35 427 211 643 1010 1011 HSA/kex2 ci) R136.HSA downstream of the HSA/kex2 leader and upstream of mature HSA.
oe Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence E
=
NO:Y NO: NO:Z NO: NO:B

,.., X
A
211 2492 pC4.IFNb(deltaM22).H Mutant full length INFbeta fused pC4 428 212 644 Native SA upstream of mature HSA. First residue of IFNI3 native, mature IFNbeta (M22) has been leader deleted.
212 2498 pC4:HSA.KGF2D60.G9 Amino acids G96 to S208 of KGF-2 fused pC4 429 213 645 1012 1013 HSA
6-S208 downstream of HSA.
n 213 2499 pC4:KGF2D60.G96- Amino acids G96 to S208 of KGF2 fused pC4 430 214 646 1014 1015 HSA

S208 :HSA upstream of mature HSA and downstream N) a, -,1 of the HSA signal peptide.
H
CA
6; 214 2501 pSAC35:scFvI006D08. BLyS antibody fused upstream of mature pSAC35 431 215 647 1016 1017 HSA/kex2 (5) Lo HSA HSA and downstream of HSA/kex2 signal I.) peptide.

a, 215 2502 pSAC35:scFvI050B11. BLyS antibody fused upstream of mature pSAC35 432 216 648 1018 1019 HSA/kex2 (5) HSA HSA and downstream of HSA/kex2 leader I.) H
sequence.
216 2513 pC4:HSA.salmoncalcito Cl through G33 of salmon calcitonin pC4 1513 1345 1681 1854 1855 HSA
nin.C1-G33 fused downstream of full length HSA.
217 2515 pC4:HDPBQ71.M1- M1 through N565 of HDPBQ71 fused pC4 1514 1346 1682 1856 1857 Native N565.HSA upstream of mature HSA

218 2529 pC4:TR1.M1- Amino acids M1 to K194 of TR1 pC4 1223 1208 1238 1253 1254 Native TR1 K194.HSA (including native signal sequence) fused 1-d upstream of mature HSA.
n 1-i 219 2530 pC4:TR1.M1- Amino acids M1 to Q193 of TR1 pC4 1224 1209 1239 1255 1256 Native TR1 Q193 .HSA (including native signal sequence) fused o t..) upstream of mature HSA.

o oe o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
o o X

220 2531 pC4:TR1.M1- Amino acids M1 to E203 of TR1 pC4 1225 1210 1240 1257 1258 Native TR1 E203 .HSA (including native signal sequence) fused upstream of mature HSA.
221 2532 pC4:TR1.M1- Amino acids M1 to Q339 of TR1 pC4 1226 1211 1241 1259 1260 Native TR1 Q339.HSA (including native signal sequence) fused upstream of mature HSA.
222 2545 pEE12.1:HDPBQ71.M1 M1 through N565 of HDPBQ71 fused pEE12.1 1515 1347 1683 Native -N565.HSA upstream of mature HSA

I.) 223 2552 pSAC35:KGF2delta33.S Amino acids S69 through S208 of KGF2 pScCHSA 1516 1348 1684 1858 1859 HSA/kex2 a, -,1 r:) H
c, 69-S208.HSA fused upstream of HSA.
co (5) 224 2553 pSAC35:HSA.KGF2delt HSA/kex2 signal peptide followed by pScNHSA 1517 1349 1685 1860 1861 HSA/kex2 co a33.S69-S208 HSA peptide followed by amino acids I.) S69 to S208 of KGF2.
a, 225 2555 pEE12.1:TR1.M1- Amino acids M1 to Q193 of TR1 pEE12.1 1227 1212 1242 Native TR1 (5) I.) Q193.HSA (including native signal sequence) fused H
upstream of mature HSA.
226 2556 pEE12.1:TR1.M1- Amino acids M1 to 1(194 of TR1 pEE12.1 1228 1213 1243 Native TR1 K194.HSA (including native signal sequence) fused upstream of mature HSA.
227 2557 pEE12.1:TR1.M1- Amino acids M1 to E203 of TR1 pEE12.1 1229 1214 1244 Native TR1 E203 .HSA (including native signal sequence) fused upstream of mature HSA.
1-d n 228 2558 pEE12.1:TR1.M1- Amino acids M1 to Q339 of TR1 pEE12.1 1230 1215 1245 Native TR1 '-Q339.HSA (including native signal sequence) fused cp o upstream of mature HSA.
t..) ' oe o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence E
NO:Y NO: NO:Z NO: NO:B
X
A
229 2571 pC4.0SCAR.R232.HSA M1 -R232 of OSCAR fused upstream of pC4 1518 1350 1686 1862 1863 Native mature HSA.
OSCAR
receptor leader 230 2580 pC4.ITNb(deltaM22,C3 IFNb fused upstream of mature HSA. The pC4 1519 1351 1687 Native 8S).HSA IFNb used in this fusion lacks the first IFI\113 residue of the mature foini of IFNb, which corresponds to M22 of SEQ ID NO:1687.
Also amino acid 38 of SEQ ID NO:1687 has been mutated from Cys to Ser.
(5) 231 2584 pC4:MPIFsp.KGF2delta MPIF signal sequence followed by A63 pC4 1520 1352 1688 1864 1865 MPIF
28.A63-S208.HSA through S208 of KGF2 followed by mature HSA.

232 2603 pC4:HSA(A14)- Modified HSA A14 leader fused upstream pC4 1521 1353 1689 Modified (5) EPO(A28-D192.G140) of mature HSA which is fused upstream HSA (A14) of A28 through D192 of EPO. Amino acid 140 of EPO is a `G'.
233 2604 pC4:HSA(S14)- Modified HSA S14 leader fused upstream pC4 1522 1354 1690 Modified EPO(A28-D192.G140) of mature HSA which is fused upstream HSA (S14) of A28 through D192 of EPO. Amino acid 140 of EPO is a 'G'.
234 2605 pC4:HSA(G14)- Modified HSA G14 leader fused upstream pC4 1523 1355 1691 Modified (1!
EPO(A28-D192.G140) of mature HSA which is fused upstream HSA (G14) g of A28 through D192 of EPO. Amino ci) acid 140 of EPO is a 'G'.
oe Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader c..4 No. ID ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o ,-, X
A
235 2606 pC4:HSA#64.KGF2D28 A63 through S208 of KGF2 fused pC4 1524 1356 1692 1866 1867 Modified .A63-S208 downstream of mature HSA and the HSA #64 modified #64 leader sequence.
236 2607 pC4:HSA#65.KGF2D28 A63 through S208 of KGF2 downstream pC4 1525 1357 1693 1868 1869 Modified .A63-S208 of mature HSA and the modified #65 HSA #65 leader sequence.
n 237 2608 pC4:HSA#66.KGF2D28 A63 through S208 of KGF2 fused pC4 1526 1358 1694 1870 1871 Modified .A63-S208 downstream of mature HSA and the HSA #66 0 I.) a, modified #66 leader sequence.
H
00"c' 238 2623 pC4:(AGVSG,14-A modified HSA A14 leader followed by pC4 1527 1359 1695 Modified Lo (5) Lo 18)HSA.HLDOU18.K2 mature HSA and amino acids K23 HSA (A14) I.) 3-R429 through R429 of HLDOU18.
leader 0 a, 239 2624 pC4:(SGVSG,14- Modified HSA S14 leader followed by pC4 1528 1360 1696 Modified 1 18)HSA.HLDOU18.K2 mature HSA and amino acids K23 to HSA (S14) (5) I.) 3-R429 R429 of HLDOU18.
leader H
240 2625 pC4:(GGVSG,14- A modified HSA 014 leader sequence pC4 1529 1361 1697 Modified 18)HSA.HLDOU18.K2 followed by mature HSA and amino acids HSA (014) 3-R429 K23 through R429 of HLDOU18.
leader 241 2630 pC4:HSA.KGF2D28.A6 Amino acids A63 to S208 of KGF-2 pC4 3-S208#2 fused to the C-terminus of HSA.
242 2631 pEE12.1:(AGVSG,14- A modified HSA Al4 leader sequence pEE12.1 1531 1363 1699 Modified 18)HSA.HLDOU18.K2 followed by mature HSA and amino acids HSA (A14) (-1 3-R429 K23 through R429 of HLDOU18.
leader '-243 2632 pEE12.1:(SGVSG,14- Modified HSA S14 leader followed by pEE12.1 1532 1364 1700 Modified ci) o 18)HSA.HLDOU18.K2 mature HSA and amino acids K23 to HSA (S14) 3-R429 R429 of HLDOU18.
leader a -Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader o No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o 1¨

X
A
244 2633 pEE12.1:(GGVSG,14- A modified HSA G14 leader sequence pEE12.1 1533 1365 1701 Modified 18)HSA.HLDOU18.K2 followed by mature HSA and amino acids HSA (G14) 3-R429 K23 through R429 of HLDOU18.
leader 245 2637 pSAC35:HSA.GCSF.T3 HSA/kex2 leader fused upstream of pScNHSA 1534 1366 1702 1874 1875 HSA/kex2 1-P207 mature HSA followed by T31 through P207 of GCSF (SEQ ID NO:1702).
n 246 2638 pPPC007:116A01.HSA scFv Il 16A01 with C-telininal HSA
pPPC007 1535 1367 1703 1876 1877 scFvI006A

fusion, where the mature form of HSA
01 I.) a, lacks the first 8 amino acids.
H
247 2647 pSAC35:T7.HSA. The T7 peptide (SEQ ID NO: 1704) of pScCHSA 1536 1368 1704 1878 1879 HSA/kex2 co (5) co Tumstatin was fused with a C-terminal I.) HSA and N terminal HSA/kex2 leader.

a, 248 2648 pSAC35:T8.HSA The T8 peptide (SEQ ID NO: 1705) of pScCHSA 1537 1369 1705 1880 1881 HSA/kex2 (5) ' Tumstatin is fused upstream to mature I.) H
HSA and downstream from HSA/kex2.
249 2649 pSAC35:HSA.T7 The T7 peptide (SEQ ID NO: 1706) of pScNHSA 1538 1370 1706 1882 1883 HSA/kex2 Tumstatin was fused with a N-terminal HSA/kex2 signal sequence.
250 2650 pSAC35:HSA.T8 The T8 peptide (SEQ ID NO: 1767) of pScNHSA 1539 1371 1707 1884 1885 HSA/kex2 Tumstatin is fused downstream to 1-d n HSA/kex2 signal sequence and mature HSA.
cp o t..) o oe vD


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader E' No. ID Vector ID ID ID ID ID Sequence o NO:Y NO: NO:Z NO: NO:B
=

X
A 1.., 251 2656 pSac35:Insulin(KR.GG Synthetic gene coding for a single-chain pScCHSA 1540 1372 1708 1886 1887 HSAikex2 G.KR).HSA insulin with HSA at C-terminus.
Contains a modified loop for processing resulting in correctly disulfide bonded insulin coupled to HSA.
252 2667 pSAC35:HSA.T1249 T1249 fused downstream of full length pSAC35 1178 1179 1180 1181 1182 HSA
HSA
(-) 253 2668 pSac35:HSA.Insulin(KR Synthetic gene coding for insulin with FL pScNHSA 1541 1373 1709 1888 1889 HSA 0 I.) .GGG.KR) HSA at N-terminus. Contains a modified a, -,1 t--µH
c) loop for processing resulting in correctly u.) c) disulfide bonded insulin coupled to HSA. (5) u.) 254 2669 pSac35:Insulin(GGG.K Synthetic gene coding for a single-chain pScCHSA 1542 1374 1710 1890 1891 HSAJkex2 I.) K).HSA insulin with HSA at C-terminus.
Contains a, a modified loop.

(5) 255 2670 pSAC35:T1249.HSA T1249 fused downstream of HSA/kex2 pSAC35 1183 1179 1180 1184 1185 HSA/kex2 "
H
leader and upstream of mature HSA.
256 2671 pSac35:HSA.Insulin(GG Synthetic gene coding for a single-chain pScNHSA 1543 1375 1711 1892 1893 HSA
G.KK) insulin with HSA at N-terminus.
Contains a modified loop for greater stability.
257 2672 pSAC35:HSA.T20 Amino terminus of T20 (codon optimized) pSAC35 1186 1187 1188 1189 1190 HSA
fused downstream of full length HSA
258 2673 pSAC35:T20.HSA Amino terminus of T20 (codon optimized) pSAC35 1191 1187 1188 1192 1193 HSA/kex2 *0 n fused downstream of HSA/kex2 leader and upstream of mature HSA.
cp 259 2700 pSAC35:HSA.GCSF.T3 C-terminal deletion of GCSF fused pSAC35 1544 1376 1712 1894 1895 HSA/kex2 2 1-R199 downstream of mature HSA.

o , oe o 1¨

Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader st), o No. ID - Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o 1¨

X
A
260 2701 pSAC35:HSA.GCSF.T3 C-terminal deletion of GCSF fused pScNHSA 1545 1377 1713 1896 1897 HSA/kex2 1-14200 downstream of mature HSA.
261 2702 pSAC35:HSA.GCSF.T3 HSA/kex2 leader followed by mature pSAC35 1194 1195 1196 1197 1198 HSA/kex2 1-L201 HSA and amino acids T31-L201 of GCSF
(corresponding to amino acids Ti to L171 of SEQ ID NO:1196).
n 262 2703 pSAC35:HSA.GCSF.A3 HSA/kex2 leader followed by mature pScNHSA 1546 1378 1714 1898 1899 HSA/kex2 6-P204 HSA and amino acids A36-P204 of I.) a, GCSF.
H
263 2714 pC4:HSASP.PTH34(2)/ PTH34 double tandem repeats fused pC4 1199 1200 1201 1202 1203 HSA
leader u.) (5) u.) HSA downstream of HSA leader (with the KEX
minus Kex I.) site deleted ¨ last 6 amino acids of the site 0 a, leader) and upstream of mature HSA.
' = 264 2724 pSAC35.sCNTF.HSA
HSA/Kex2 fused to CNTF, and then pSAC35 1547 1379 1715 1900 1901 HSA/kex2 (5) I.) fused to mature HSA.
H
265 2725 pSAC35:HSA.sCNTF HSA/Kex2 fused to mature HSA and pSAC35 1548 1380 1716 1902 1903 HSA/kex2 then to CNTF
266 2726 pSac35.INV.GYGinsuli Synthetic gene coding for a single-chain pSAC35 1549 1381 1717 1904 1905 Invertase n.HSA insulin with HSA at C-terminus. The signal peptide of invertase is used for this construct.
Iv 267 2727 pSac35.INV.GYGinsuli Synthetic gene coding for a single-chain pSAC35 1550 1382 1718 1906 1907 invertase n n(delF1).HSA insulin with HSA at C-terminus.
Construct uses the invertase signal peptide cp o and is lacking the first amino acid (F) of t..) o mature human insulin.
ce o 1¨

, Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B

X
A 1.., 268 2749 pEE12.1.0SCAR.R232. Amino acids M1 through R232 of pEE12.1 1551 1383 1719 1908 1909 Native HSA OSCAR fused upstream of mature HSA.
OSCAR
leader 269 2784 pSAC35:Insulin(GYG)- Synthetic gene coding for a single-chain pSAC35 1552 1384 1720 1910 1911 invertase HSA codon optimized insulin with HSA at C-terminus.
270 2789 pSAC35:Insulin(GGG). Synthetic gene coding for a single-chain pSAC35 1553 1385 1721 1912 1913 invertase n HSA (codon optimized) insulin with HSA at C-terminus.
, 271 2791 pEE12.1:HSAsp.PTH34 Parathyroid hormone is fused in tandem pEE12.1 1554 1386 1722 HSA leader 0 I.) a, (2X).HSA and upstream of mature HSA and minus Kex H
I-CA
0 downstream from HSA signal peptide site (5) t.) u.) (with the KEX site deleted ¨ last 6 amino I.) acids of the leader) a, 272 2795 pC4:HSA(A14)- The mature form of IFNb is fused to the pC4 1555 1387 1723 Modified 1 IFNb.M22-N187 C-terminus of HSA, which contains an HSA (A14) (5) , I.) modified signal peptide, designed to H
improve processing and homogeneity.
273 2796 pC4:HSA(S14)- The mature form of IFNb is fused to the pC4 1556 1388 1724 Modified IFNb.M22-N187 C-terminus of HSA, which contains a HSA (S14) modified signal peptide, designed to improve processing and homogeneity.
274 2797 pC4:HSA(G14)- The mature form of IFNb is fused to the pC4 1557 1389 1725 Modified IFNb.M22-N187 C-terminus of HSA, which contains an HSA (G14) 'A
modified signal peptide.
275 2798 pSAC35:Somatostatin(S A 14 amino aid peptide of Somatostatin pScCHSA 1558 1390 1726 1914 1915 HSA/kex2 i 14).HSA fused downstream of HSA/kex2 leader t..) and upstream upstream of mature HSA.
oe vD


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID
Vector'a ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
' 1.., X
A
276 2802 pSAC35:GLP-1(7- GLP-1(7-36(A80)) (SEQ ID NO:1808) is pScNHSA 1559 1391 1727 HSA/kex2 36(A8G)),IP2.HSA fused downstream from the HSA/kex2 leader sequence and upstream from the intervening peptide-2 of proglucagon peptide and upstream from mature HSA.
277 2803 pSAC35:GLP-1(7- GLP-1(7-36(A8G)) (SEQ ID NO:1808) is pScCHSA 1231 1216 1246 1261 1262 HSA/kex2 r) 36(A8G))x2.HSA tandemly repeated and fused downstream of the HSA/kex2 signal sequence, and I.) a, upstream of mature HSA.
H
' c) 278 2804 pSAC35:coGLP-1(7-GLP-1(7-36(A8G)) (SEQ ID NO:1808) is pScCHSA 1232 1217 1247 1263 1264 HSA/kex2 u.) (5) (4.) 36(A8G))x2.HSA tandemly repeated and fused downstream u.) I.) of the HSA/kex2 signal sequence, and a, upstream of mature HSA.

279 2806 pC4:HSA#65.salmoncal Modified HSA leader #65 followed by pC4 1560 1392 1728 1916 1917 Modified (5) citonin.C1-G33 mature HSA and C1-G33 of salmon HSA #65 I.) H
calcitonin.
280 2821 pSac35.delKex2.Insulin( Synthetic gene coding for a single-chain pScCHSA 1561 1393 1729 Modified GYG).HSA insulin with HSA at C-terminus. The kex2 HSA/kex2, site has been deleted from the HS.A/KEX2 lacking the signal peptide.
Kex2 site.
281 2822 pSac35.alphaMF.Insulin Synthetic gene coding for a single-chain pSAC35 1562 1394 1730 1920 1921 AfFa-1 (GYG).HSA insulin with HSA at C-terminus. The 1-d n signal peptide of alpha mating factor (MFV-1) is used for this construct.
cp o 282 2825 pSAC35:HSA.Somatost 14 amino acid peptide of Somatostatin pScNHSA 1563 1395 1731 1922 1923 HSA/kex2 t-) .1-.
atin(S14) was fused downstream of HSA/kex2 =
oe leader and mature HSA.
o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader F,3 No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B
X
A
283 2830 pSAC35:S28.HSA 28 amino acids of somatostatin fused pScCHSA 1564 1396 1732 1924 1925 HSA/kex2 downstream of HSA/kex2 leader and upstream of mature HSA.
284 2831 pSAC35:HSA.S28 28 amino acids of somatostatin fused pScNHSA 1565 1397 1733 1926 1927 HSA/kex2 downstream of HSA/kex2 leader and mature HSA.
285 2832 pSAC35:Insulin.HSA Long-acting insulin peptide fused pScCHSA 1566 1398 1734 1928 1929 invertase (yeast codon optimized) upstream of mature HSA.

286 2837 pSAC35:CKB1.K21- K21-N93 of CKB1 (fragment shown as pScCHSA 1567 1399 1735 1930 1931 HSA/kex2 N93:HSA K2 to N74 of SEQ ID NO:1735) fused (5) downstream of the HSA/kex2 leader and upstream of mature HSA.

287 2838 pSAC35:CKB1.T22- T22-N93 of CKB1 (fragment shown as T3 pScCHSA 1568 1400 1736 1932 1933 HSA/kex2 N93:HSA to N74 of SEQ ID NO:1736) fused (5) downstream of the HSA/kex2 leader and upstream of mature HSA.
288 2839 pSAC35:CKB1.E23-. E23-N93 of CKB1 (fragment shown as E4 pScCHSA 1569 1401 1737 1934 1935 HSA/kex2 N93:HSA to N74 of SEQ ID NO:1737) fused downstream of the HSA/kex2 leader and upstream of mature HSA.
289 2840 pSAC35:CKB1.S24- S24-N93 of CKB1 (fragment shown as S5 pScCHSA 1570 1402 1738 1936 1937 HSA/kex2 N93:HSA to N74 of SEQ ID NO:1738) fused 1-d downstream of the HSA/kex2 leader and upstream of mature HSA.
oe =
Table 2 _______________________________________________________________________________ _______________________________________ 0 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a No. ID Vector ID ID ID ID ID Sequence Cf NO:Y NO: NO:Z NO: NO:B
X
A
290 2841 pSAC35:CKB1.S25- S25-N93 of CKB1 (fragment shown as S6 pScCHSA 1571 1403 1739 1938 1939 HSA/kex2 N93:HSA to N74 of SEQ ID NO:1739) fused downstream of the HSA/kex2 leader and upstream of mature HSA.
291 2842 pSAC35:CKB1.S26- S26-N93 of CKB1 (fragment shown as S7 pScCHSA 1572 1404 1740 1940 1941 HSA/kex2 N93:HSA to N74 of SEQ ID NO:1740) fused downstream of the HSA/kex2 leader and upstream of mature HSA.

292 2843 pSAC35:CKB1.R27- R27-N93 of CKB1 (fragment shown as pScCHSA 1573 1405 1741 1942 1943 HSA/kex2 N93:HSA R8 to N74 of SEQ ID NO:1741) fused (5) downstream of the HSA/kex2 leader and upstream of mature HSA.

293 2844 pSAC35:CKB1.P29- P29-N93 of CKB1 (fragment shown as pScCHSA 1574 1406 1742 1944 1945 HSA/kex2 N93:HSA P10 to N74 of SEQ ID NO:1742) fused (5) downstream of the HSA/kex2 leader and upstream of mature HSA.
294 2845 pSAC35:CKB1.Y30- Y30-N93 of CKB1 (fragment shown as pScCHSA 1575 1407 1743 1946 1947 HSA/kex2 N93:HSA Y1 1 to N74 of SEQ ID NO:1743) fused downstream of the HSA/kex2 leader and upstream of mature HSA.
295 2849 pC4.MPIFsp.CKB1.G28 G28-N93 of CKB1 (fragment shown as pC4 1576 1408 1744 1948 1949 MPIF
-N93 .HSA G9 to N74 of SEQ ID NO:1744) fused 1-d downstream of the MPIF signal peptide and upstream of mature HSA.
296 2872 pSAC35:HSA.IFNaA(C This construct contains a hybrid foim of pSAC35 1309 1310 1311 1312 1313 HSA/kex2 1-Q91)/ D(L93-E166) IFNaA and IFNaD fused downstream of oe mature HSA.

Table 2 ' Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader E..
No. ID Vector ID ID ID ID ID Sequence E
=
NO:Y NO: NO:Z NO: NO:B

.
1.., X
A
297 2873 pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 1314 1315 1316 1317 1318 HSA/kex2 1-Q91)/ B(L93-E166) IFNaA and IFNaB fused downstream of mature HSA.
298 2874 pSAC35:HSA.IFNaA(C This construct contains a hybrid fowl of pSAC35 1319 1320 1321 1322 1323 HSA/kex2 1-Q91)/ F(L93-E166) IFNaA and IFNaF fused downstream of mature HSA.
r) 299 2875 pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 1324 1325 1326 1327 1328 HSA/kex2 1Q-62)/D(Q64-E166) IFNaA and 1FNaD fused downstream of N) a, mature HSA.
H
CA
8 300 2876 pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 1329 1330 1331 1332 1333 HSA/kex2 (5) u.) 1-Q91)/ D(L93-E166); IFNaA and 1FNaD fused downstream of I.) R23K,A113V mature HSA.

a, ' 301 2877 pSAC35:KT.Insulin.HS Killer toxin signal peptide fused to pScCHSA 1577 1409 1745 1950 1951 Killer toxin 0 A synthetic gene coding for a single-chain (5) I.) insulin with C-terminal HSA
H
302 2878 pSAC35:AP.Insulin.HS Acid phospatase signal peptide fused to pSAC35 1578 1410 1746 Acid A synthetic gene coding for a single-chain phosphatas insulin with C-terminal HSA.
e 303 2882 pSac35.alphaMFprepro.I MFa-1 prepro signal followed by GYG
pSAC35 1579 1411 1747 MFa-1 nsulin(GYG).HSA insulin followed by mature HSA.
304 2885 pSac35.alphaMFpreproE Yeast MFa-1 prepro signal followed by pSAC35 1580 1412 1748 Yeast 1-d EA.Insulin(GYG).HSA GYG insulin follwed by mature HSA.
MFa-1 n 1-i 305 2886 pSAC35:HSA.GCSF.P4 HSA/kex2 signal peptide followed by pSAC35 1581 1413 1749 1952 1953 HSA/kex2 -----0-P204 mature HSA followed by GCSF (P40-cp o P204).
t..) .1-.
o oe o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader F...4 No. ID Vector ID ID ID ID ID Sequence E
NO:Y NO: NO:Z NO: NO:B
X
A
306 2887 pSAC35:HSA.GCSF.P4 HSA/kex2 signal peptide followed by pSAC35 1582 1414 1750 1954 1955 HSA/kex2 0-L201 mature HSA followed by GCSF (P40-L201).
307 2888 pSAC35:HSA.GCSF.Q4 HSA/kex2 signal peptide followed by pSAC35 1583 1415 1751 1956 1957 HSA/kex2 1-L201 mature HSA followed by GCSF (Q41-L201).
308 2889 pSAC35:HSA.GCSF.Q4 HSA/kex2 signal peptide followed by pSAC35 1584 1416 1752 1958 1959 HSA/kex2 1-P204 mature HSA followed by GCSF (Q41-P204).
309 2890 pC4.HSA.GCSF.T31- HSA/kex2 signal peptide followed by pC4 1585 1417 1753 1960 1961 HSA/kex2 (5) P204 mature HSA followed by GCSF (T31-P204).

310 2891 pGAP.alphaMF.Insulin( Synthetic gene coding for a single-chain pYPGaf 1586 1418 1754 1962 1963 HSA/kex2 GYG).HSA insulin with HSA at C-terminus. The (5) signal peptide of HSA/kex2 is used for this construct.
311 2897 pGAP.Insulin(KR.GGG. Long-acting insulin analog using a pYPGaf 1587 1419 1755 1964 1965 HSA/kex2 KR).HSA synthetic gene coding for a single-chain insulin with HSA at C-terminus. Contains a modified loop for processing resulting in correctly disulfide bonded insulin coupled to HSA
1-d 312 2900 pSAC:GLP-1(7- GLP-1(7-36) is tandemly repeated and pScCHSA 1233 1218 1248 1265 1266 HSA/kex2 36)x2.HSA then fused downstream of the HSA/kex2 signal sequence and upstream of mature EISA.
oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
313 2901 pSAC35:IL22.A18- Amino acids A18-P202 of IL22 fused pSAC35 1588 1420 1756 1966 1967 HSA/kex2 P202.HSA downstream of HSA/kex2 leader and upstream of mature HSA.
314 2902 pSAC35: A 14 amino acid peptide of Somatostatin, pScCHSA 1589 1421 1757 1968 1969 HSA/kex2 Somatostatin(S14(A- an inhibitor of growth homione, G)).HSA synthesized as a C-teiminal HSA fusion.
Somatostatin has an alanine to glycine change at amino acid 1 of SEQ ID

NO:1757.

2903 pSAC35:HSA.A18- Amino acids A18-P202 of IL22 fused pSAC35 1590 1422 1758 1970 1971 HSA
(5) co P202.IL22 downstream of full length HSA.
316 2904 pSAC35:GLP-1(9- Amino acids E100 to R127 of pScCHSA 1234 1219 1249 1267 1268 HSA/kex2 0 36).GLP-1(7-36).HSA preproglucagon (SEQ ID NO:1249) (hereinafter, this particular mutant is (5) referred to as GLP-1(9-36)) is fused downstream from the HSA/kex2 signal sequence and upstream from GLP-1(7-36), and mature HSA.
317 2908 pSAC35:HSA.HCE1P8 Mature HSA fused downstream of the pSAC35 1591 1423 1759 1972 1973 HSA/kex2 0 HSA/kex2 leader and upstream of HCE1P80.
318 2909 pSAC35:HSA.HDRMI8 Mature HSA fused downstream of the pSAC35 1592 1424 1760 1974 1975 HSA/kex2 2 HSA/kex2 leader sequence and upstream of HDRMI82.
319 2910 pSAC35:HSA.RegIV Mature HSA fused downstream of the pSAC35 1593 1425 1761 1976 1977 HSA/kex2 HSA/kex2 leader sequence and upstream oo of RegIV.

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
320 2915 pC4:HSA#65.humancalc Modified HSA leader #65 followed by pC4 1594 1426 1762 1978 1979 Modified itonin.C1-G33 mature HSA and C98 through G130 of HSA #65 SEQ ID NO: 1762.
321 2930 pC4.MPIF.Insulin(GYG Insulin is downstream of an MPIF signal pC4 1595 1427 1763 1980 1981 MPIF
).HSA peptide and upstream of mature HSA.
322 2931 pC4.HSA.Insulin(GYG) Synthetic gene coding for a mature single- pC4 1596 1428 1764 1982 1983 Modified chain insulin fused downstream of the HSA (A14) modified HSA A14 leader and mature leader 0 HSA.
323 2942 pSac35.TA57.Insulin(G The TA57 Propeptide fused to a single pScNHSA 1597 1429 1765 1984 1985 TA57 (5) YG).HSA chain insulin (GYG), and then mature propeptide HSA.

324 2943 pSAC35:HSA.T7.T7.T7 Dimer construct- HSA/kex2 leader pScNHSA 1598 1430 1766 1986 1987 HSA/kex2 4-L98 followed by mature HSA followed by two (5) copies of T7 peptide (SEQ ID NO:1766) of Tumstatin.
325 2944 pSAC:HSA.T8.T8.K69- HSA/kex2 leader followed by mature pScNHSA 1599 1431 1767 1988 1989 HSA/kex2 S95 HSA followed by two copies of T8 peptide (SEQ ID NO: 1767) of Tumstatin 1-d oe Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader st), No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
326 2945 pSAC35:GLP-1(7- Amino acids H98 to R127 of pScCHSA
1235 1220 1250 1269 1270 HSA/kex2 36(A8S)).GLP-1(7- preproglucagon (SEQ ID NO:1250) is 36).HSA mutated at position 99 from alanine to senile (hereinafter, this particular mutant is referred to as GLP-1(7-36(A8S)), which is fused downstream from the HSA/kex2 signal sequence and upstream from GLP-1(7-36), and mature HSA.

327 2946 pSAC:T1249(x2).HSA This dimer represents the wild type pScCHSA 1600 1432 1768 1990 1991 HSA/kex2 sequence for T1249. Both dimers have (5) been yeast codon optimized. The second dimer was optimized to be different from the first (at the wobble position) to ensure good amplification. Construct has the (5) HSA/kex2 leader followed by T1249 dimer followed by mature HSA.
328 2947 pSAC:CKb- Invertase signal peptide followed by pSAC35 1601 1433 1769 1992 1993 invertase 188(x2).HSA amino acids G28-N93 of full length C1(131 (SEQ IDNO:1769), tandemly repeated, followed by mature HSA.
329 2964 pSAC35:GLP-1(7- GLP-1(7-36) is tandemly repeated as a pSAC35 1236 1221 1251 1271 1272 HSA/kex2 36)x2.HSA dimer and fused downstream from the 1-d HSA/kex2 leader sequence and upstream from mature HSA.
330 2965 pC4:MPIFspP.PTH(1- MPIF signal peptide followed by 34 pC4 1602 1434 1770 1994 1995 MPIF
34).HSA amino acids of PTH followed by mature oe HSA.

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B

X

331 2966 pEE12:MPIFsp.PTH(1- MPIF signal peptide followed by 34 PEE12.1 1603 1435 1771 1996 1997 MPIF
34).HSA amino acids of PTH followed by mature HSA.
332 2982 pSAC35:GLP-1(7- GLP-1(7-36(A8G)) (SEQ ID NO:1808) is pScCHSA 1237 1222 1252 1273 1274 HSA/kex2 36(A8G).GLP-1(7- fused downstream from the HSA/kex2 36).HSA signal sequence and upstream from GLP-1(7-3 6) and mature HSA.
n 333 2983 pC4.HSA.Growth Modified (A14) HSA leader followed by pC4 1604 1436 1772 1998 1999 Modified 0 I.) Hormone.F27-F-217 mature HSA followed by F27 through HSA (A14) a, -,1 H
!--, ,--, F217 of growth hormone (corresponding u.) (5) ,¨ to amino acids Fl to F191 of SEQ ID
u.) I.) NO:1772).

334 2986 pSac35.y3SP.TA57PP.I The TA57 Propeptide fused to a single pScCHSA 1605 1437 1773 2000 2001 TA57 a, nsulin(GYG).HSA chain insulin (GYG), and then mature propeptide (5) H
335 3025 pSAC35:INU.Insulin.H Inulinase signal peptide is fused pScCHSA 1606 1438 1774 2002 2003 inulinase SA upstream of single chain insulin (GYG) and HSA.
336 3027 pSAC35:INV.GLP-1(7- Invertase signal peptide followed by GLP- pSAC35 1607 1439 1775 2004 2005 invertase 36A8G)x2.HSA 1(7-36(A8G)) (SEQ ID NO:1808) tandemly repeated as a dimer, followed by mature HSA.
1-d n 337 3028 pSAC35:INV.GLP-1(7- Invertase signal peptide followed by GLP- pSAC35 1608 1440 1776 2006 2007 invertase 36(A8G)).GLP-1(7- 1(7-36(A8G)) (SEQ ID NO:1808), then cp 36).HSA GLP-1(7-36(A8G)), and then mature o t..) .1-.
HSA.
=
oe o 1¨

=
Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID
VectorO' ID ID ID ID ID Sequence o o NO:Y NO: NO:Z NO: NO:B
=

X

338 3045 pSAC35:DeltaKex.GLP- HSA/kex2 signal sequence, minus the last pSAC35 1609 1440 1776 2008 2009 HSA/kex2 1(7-36A8G)x2.HSA six amino acids of the leader, is fused to last six GLP-1(7-36(A8G)) (SEQ ID NO:1808) amino which is tandemly repeated as a dimer, acids followed by mature HSA.
339 3046 pSAC35:Delta HSA/kex2 signal sequence, minus the last pSAC35 1610 1440 1776 2010 2011 HSA/kex2 Kex.GLP-1(7- six amino acids of the leader, is fused to last six n 36A8G).GLP-1(7- GLP-1(7-36(A8G)) (SEQ ID NO:1808), amino 0 I.) 36).HSA GLP-1(7-36), and mature HSA.
acids a, -,1 H
340 3047 pSAC35: HSA.Tum5 Full length HSA fused to the Tum5 pScNHSA 1611 1443 1779 2012 2013 HSA Lo Lo peptide (SEQ ID NO:1779) of Tumstatin.
I.) 341 3048 pSAC35: Tum5.HSA. The Tum5 peptide (SEQ ID NO:1780) of pScCHSA 1612 1444 1780 2014 2015 HSA/kex2 0 Tumstatin is fused to HSA and HSA/kex2 a, leader.
(5) 342 3049 pC4.HSA.HCE1P8O.D9 Amino acids D92 to L229 of HCE1P80 pC4 1613 1445 1781 2016 2017 HSA I.) H
2-L229 are fused downstream of the full length HSA.
343 3050 pC4.HSA.HCE1P80.A2 Amino acids A20-L229 of HCE1P80 are pC4 0-L229 fused downstream of the full length human HSA
344 3051 pSAC35.HSA.HCE1P8 Amino acids D92 to L229 of HCE1P80, a pSAC35 1615 1447 1783 2020 2021 HSA
0.D92-L229 member of the Clq family of proteins, are 1-d n fused downstream of the full length human HSA
cp =
345 3052 pSAC35.HSA.HCE1P8 Amino acids A20-L229 of HCE1P80 are pSAC35 1616 1448 1784 2022 2023 HSA t..) .1-.
0.A20-L229 fused downstream of the full length =
oe human HSA
vD


=
' Table 2 _______________________________________________________________________________ _________________________________________ =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader .._,..?
o No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B o 1.., X
A
346 3053 pC4.HSA.HDALV07.K The globular domain of adiponectin pC4 101-N244 (amino acids K101-N244) has been inserted downstream of full length human HSA
347 3055 pSAC35.HSAHDALVO Full length HSA followed by amino acids pSAC35 1618 1450 1786 2026 2027 HSA
7(GD) K101-N244 of HDALV07(GD)/
n Adiponectin.
, 348 3056 pSAC35.HSA.HDALVO Full length HSA followed by amino acids pSAC35 1619 1451 1787 2028 2029 HSA I.) a, 7.MP Q18 to N244 of HDALV07.
H
CA
349 3066 pSAC35:CKB- Invertase signal peptide followed by pScCHSA 1620 1452 1788 2030 2031 invertase (5) 1d8.GLP-1(7-36).HSA amino acids G28-N93 of full length CKf31 I.) (SEQ IDNO:1788), followed by GLP-1(7-a, , 36), followed by mature HSA.

350 3069 pSAC35:INU.GLP-1(7- The inulinase signal sequence is fused to pSAC35 1621 1453 1789 2032 2033 inulinase (5) I.) 36(A8G))x2.HSA GLP-1(7-36(A8G)) (SEQ ID NO:1808), H
which is tandemly repeated as a dimer and fused to mature HSA.
351 3070 pSAC35:KT.GLP-1(7- GLP-1(7-36(A8G)) (SEQ ID NO:1808) is pSAC35 1280 1281 1282 1283 1284 Killer toxin 36(A8G))x2.HSA tandemly repeated as a dimer and fused upstream from mature HSA and downstream from the killer toxin signal 1-d sequence. -n 352 3071 pSAC35:MAF.GLP-1(7- The yeast mating factor a-1 (hereinafter pSAC35 1622 1454 1790 2034 2035 MFa-1 36(A80))x2.HSA MFa-1) signal sequence is fused to cp o tandemly repeated copies of GLP-1(7-i..) 36(A8G)) (SEQ ID NO:1808), which are o oe o fused to mature HSA.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader F...4 o No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o --.1 1.., X
A
353 3072 pSAC35:AP.GLP-1(7- The acid phosphatase signal sequence is pSAC35 1623 1455 1791 2036 2037 Acid 36(A8G))x2.HSA fused to tandemly repeated copies of phosphatas GLP-1(7-36(A8G)) (SEQ ID NO:1808), e which are fused to mature HSA.
354 3085 pSAC35:MAF.GLP-1(7- The yeast mating factor a-1 (hereinafter pSAC35 1624 1456 1792 2038 2039 MFa-1 36(A8G)).GLP-1(7- MFa-1) signal sequence is fused to GLP-n 36).HSA 1(7-36(A.G)) (SEQ JD NO:1808), GLP-1(7-36), and mature HSA.

I.) a, 355 3086 pSAC35:lNU.GLP-1(7- The inulinase signal sequence is fused to pSAC35 1625 1457 1793 2040 2041 inulinase H
,¨ 36(A8G)).GLP-1(7- GLP-1(7-36(A8G)) (SEQ ID NO:1808), u.) (5) V. 36).HSA GLP-1(7-36), and mature HSA.
u.) I.) 356 3087 pSAC35:AP.GLP-1(7- The acid phosphatase signal sequence is pSAC35 1626 1458 1794 2042 2043 Acid 0 a, 36(A8G)).GLP-1(7- fused to GLP-1(7-36(A8G)) (SEQ ID
phosphatas 0 36).HSA NO:1808), GLP-1(7-36), and mature e (5) , I.) HSA.
H
357 3088 pSAC35.HSA.C-Peptide HSA/kex2 signal peptide, followed by pSAC35 1627 1459 1795 2044 2045 HSA/kex2 HSA, followed by the C-Peptide sequence.
358 3105 pSAC35:INV.t9HCC- Invertase signal peptide followed by pSAC35 1628 1460 1796 2046 2047 invertase 1.G28-N93:spc.HSA amino acids G28 to N93 of HCC-1 fused upstream of a spacer and mature HSA.
359 3106 pSACHSA.HCBOG68 mature HCBOG68 fused downstream of pSAC35 1629 1461 1797 HSA/kex2 A
mature HSA and the HSA/kex2 leader sequence.
cp o 360 3108 pSAC35HSA.PYY Mature PYY fused downstream of mature pSAC35 1630 1462 1798 HSA/kex2 "
.1-.
HSA and the HSA/kex2 leader.
oe o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader ca.) No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B



X
A
361 3109 pSAC35HSA.PYY3-36 HSA/kex2 leader followed by mature pSAC35 1631 1463 1799 HSA/kex2 HSA and then PYY3-36 (SEQ ID
NO:1799).
362 3117 pC4:PYY3-36/HSA HSA leader followed by PYY3-36 (SEQ pC4 ID NO:1800) and mature HSA.
363 3118 pSAC35:PYY3-36/HSA HSA/kex2 leader followed by PYY3-36 pSAC35 1633 1465 1801 2050 2051 HSA/kex2 n (SEQ ID NO:1801) and mature HSA.
364 3119 pSAC35:BNP/HSA HSA/kex2 leader followed by BNP and pSAC35 1634 1466 1802 2052 2053 HSA/kex2 0 I.) a, mature HSA.
H, '",---: 365 3124 pSAC35:INV.CKB1.P2 Invertase signal peptide followed by pSAC35 1635 1467 1803 2054 2055 invertase u.) (5) u.) 9-N93:HSA amino acids 29 to 93 of full length I.) ckbetal fused to N-terminus of HSA.

366 3125 pSAC35:INV.CKb- Invertase signal peptide followed by pSAC35 1636 1468 1804 2056 2057 invertase 1 1.R27-N93:HSA amino acids 27 to 93 of full length (5) , I.) ckbetal fused to N-terminus of HSA.
H
367 3133 pSac35.ySP.TA57PP.Ins Variant TA57 propeptide leader followed pSAC35 1637 1469 1805 2058 2059 TA57 ulin(GYG).HSA by single chain insulin, followed by variant 1 mature HSA.
368 3134 pSac35.ySP.TA57PP+S. Variant TA57 propeptide leader followed pSAC35 1638 1470 1806 2060 2061 TA57 Insulin(GYG).HSA by single chain insulin, followed by variant 2 mature HSA.
369 3139 pSAC35:INV.CKB1.G2 Invertase signal peptide followed by pSAC35 1639 1471 1807 2062 2063 invertase A
8-N93.DAHK.HSA amino acids 028-N93 of full length (see, e.g, SEQ IDNO:1788), followed by a cp o 16 amino acid linker derived from the N-t..) .1-.
terminus of HSA, followed by mature oe o HSA.
1--, Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a No. ID Vector ID ID ID ID ID Sequence o NO:Y NO: NO:Z NO: NO:B
o X
A 1.., 370 3140 pSAC35:GLP1(mut)DA GLP-1(7-36(A8G)) (SEQ ID NO:1808) is pSAC35 1640 1472 1808 2064 2065 HSA/kex2 HK.HSA linked to mature HSA by a 16 amino acid linker derived from the N-terminus of HSA. The HSA/kex2 signal sequence is used.
371 3141 pSAC35:Wntl0b/HSA HSA/kex2 leader followed by amino pSAC35 1641 1473 1809 2066 2067 HSA/kex2 acids N29 to 1(389 of Wntl Ob followed by mature HSA.

N
372 3142 pSAC35:Wnt11/HSA HSA/kex2 leader followed by mature pSAC35 1642 1474 1810 2068 2069 HSA/kex2 a, -,1 ,-. Wntl 1 followed by mature HSA.
H
CA
I--, c: 373 3143 pSAC35:herstatin/HSA HSA/kex2 leader followed by amino pSAC35 1643 1475 1811 2070 2071 HSA/kex2 co I.) acids T23 to G419 of herstatin followed by mature HSA.
a, 374 3144 pSAC35:adrenomedul1in HSA/kex2 leader followed by amino pSAC35 1644 1476 1812 2072 2073 HSA/kex2 (5) (27-52)/HSA acids 27-52 of adrenomedullin followed "
, by mature HSA.
375 3149 pSAC35.HSA.C-peptide Full length HSA fused to amino acids E7 pSAC35 1645 1477 1813 2074 2075 HSA
tandem to Q37 of SEQ ID NO:1813, tandemly repeated.
376 3152 pSAC35:INV.CKB1.Me Invertase signal peptide followed by a pSAC35 1646 1478 1814 2076 2077 invertase t.R27-N93.HSA Met, followed by amino acids R27-N93 of full length CK131, followed by mature 1-d n HSA.
377 3153 pSAC35:INV.CKB1.Me Invertase signal peptide followed by a pSAC35 1647 1479 1815 2078 2079 invertase c7, t.526-N93.HSA Met, followed by amino acids S26-N93 of o w full length CI(131, followed by mature .1-.
o oe HSA.
o 1¨

Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: ' NO:B

X
A 1.., 378 3154 pSAC35:INV.CKB1.Me Invertase signal peptide followed by a pSAC35 1648 1480 1816 2080 2081 invertase t.S25-N93.HSA Met, followed by amino acids S25-N93 of full length CK131, followed by mature HSA.
379 3155 pSAC35:INV.CKB1.Me Invertase signal peptide followed by a pSAC35 1649 1481 1817 2082 2083 invertase t.G28-N93.HSA Met, followed by amino acids G28-N93 of full length cKpi, followed by mature n HSA.

I.) 380 3156 pSAC35:INV.CKB1.Me Invertase signal peptide followed by a pSAC35 1650 1482 1818 2084 2085 invertase a, -,1 H
,-- t.P29-N93.HSA Met, followed by amino acids P29-N93 of u.) (5) )-7-1 full length CK131, followed by mature u.) I.) HSA.

381 3163 pSAC35:HSA.hGH HSA/kex2 leader fused upstream of pSAC35 1303 1304 1305 HSA/kex2 a, mature HSA and 191 amino acids of hGH.
(5) 382 3165 pSAC35:HSA.IFNa HSA fused upstream of IFNa and pSAC35 1300 1301 1302 HSA/kex2 I.) H
downstream of the HSA/kex2 leader.
also named CID 3165, pSAC35:HSAINFa 383 3166 pC4:MPIF1.A22- Amino acids A49 to N120 of MPIF (SEQ
pC4 1651 1483 1819 2086 2087 MPIF
N93.HSA ID NO:1821) is fused downstream of MPIF signal peptide and upstream of mature HSA.
1-d n 384 3167 pC4:HSA.MP1F1.D45- Full length HSA followed by amino acids pC4 1652 1484 1820 2088 2089 HSA
N120 D45 through N120 of MPIF.
cp 385 3168 PC4:MPIF-1.HSA Amino acids D45 through N120 of MPIF
pC4 1653 1485 1821 2090 2091 MPIF t..) fused downstream downstream of the MPIF signal =
oe vD
sequence and upstream of mature HSA.


Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence E
NO:Y NO: NO:Z NO: NO:B

1¨, X
A
386 3169 pSAC35:KT.CKB1.G28 Killer toxin signal sequence fused pSAC35 1654 1486 1822 Killer toxin -N93 .HSA upstream of amino acids G28 through N93 of CKB1 (fragment shown as amino acids G1 to N66 of SEQ ID NO:1822) and mature HSA.
387 3170 pSAC35:KT.HA.CICB1. Killer toxin signal sequence followed by pSAC35 1655 1487 1823 Killer toxin G28-N93.HSA HA dipeptide and amino acids G28 n through N93 of CKB1 (fragment shown I.) a, as amino acids G1 to N66 of SEQ ID
H
,--, NO:1823) and mature HSA.
Lo ,--, (5) m 388 3171 pSAC35:sCNTF(M1- C-terminal deletion of CNTF (amino acids pSAC35 1656 1488 1824 2092 2093 HSA/kex2 Lo I.) G185):HSA M1 through G185), fused upstream of mature HSA and codon optimized for a, expression in yeast. HSA/kex2 signal (5) I.) sequence is used.
389 3172 pSAC35:HSA: HSA/kex2 signal sequence followed by pSAC35 1657 1489 1825 2094 2095 HSA/kex2 5CNTF(M1-G185) mature HSA and M1 through G185 of CNTF.
390 3184 pC4:HSA.NOGOR.C27- Full length HSA followed by amino acids pC4 1658 1490 1826 2096 2097 HSA
C309 C27 to C309 of the NOGO receptor.
391 3185 pC4.NOGOR.M1- Amino acids Ml-C309 of NOGO pC4 1659 1491 1827 2098 2099 Native C309.HSA receptor fused upstream of mature HSA.
NOGO 1-d n receptor 392 3194 pC4:HSA(A14)- Codon optimized EPO(A28-D192.G140) pC4 1660 1492 1828 2100 2101 modified c7, EPO(A28- fused downstream of mature HSA with a HSA (A14) 2 D192.G140)codon opt modified HSA (A14) signal sequence.
o oe vD
1--, =
Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader a' No. ID Vector ID ID ID ID ID Sequence I
NO:Y NO: NO:Z NO: NO:B

X

393 3195 pC4:HSA(S14)- Codon optimized EPO(A28-D192.G140) pC4 1661 1493 1829 2102 2103 modified EPO(A28- fused downstream of mature HSA and a HSA (S14) D192.G140)codon opt modified HSA (S14) signal sequence.
394 3196 pC4:HSA(G14)- Codon optimized EPO(A28-D192.G140) pC4 1662 1494 1830 2104 2105 modified EPO(A28- fused downstream of mature HSA with a (G14) D192.G140)codon opt modified (G14) HSA signal sequence.
395 3197 pC4.MPIF.Insulin(EAE) A single-chain insulin is downstream of pC4 1663 1495 1831 MPIF n .HSA the MPIF signal peptide and upstream of I.) mature human HSA.
a, -,1 H
396 3198 pSac351NV.insulin(EA Single-chain insulin is downstream of the pSAC35 1664 1496 1832 invertase Lo (5) E).HSA invertase signal peptide and upstream of Lo I.) mature human HSA

397 3202 pSAC35:API.d8CKbl/H HSA/kex2 leader followed by amino acids pSAC35 1665 1497 1833 2106 2107 HSA/kex2 a, SA "API" followed by d8CKbl and mature (5) HSA. The sequence of delta 8 for CKB1 I.) H
is shown in SEQ ID NO:1833.
398 3203 pSAC35:ASL.d8CKb1/ HSA/kex2 leader followed by amino acids pSAC35 1666 1498 1834 2108 2109 HSA/kex2 HSA "ASL" followed by d8CKbl and mature HSA.
399 3204 pSAC35:SPY.d8CKb1/ HSA/kex2 leader followed by amino acids pSAC35 1667 1499 1835 2110 2111 HSA/kex2 HSA "SPY" followed by d8CKbl and mature HSA.
1-d n 400 3205 pSAC35:MSPY.d8CKb HSA/kex2 leader followed by amino acids pSAC35 1668 1500 1836 2112 2113 HSA/kex2 1/HSA "MSPY" followed by d8CKbl and mature cp o HSA.
t..) .1-.
=
oe o 1¨

Table 2 =
Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader E' No. ID Vector ID ID ID ID ID Sequence `1,--o NO:Y NO: NO:Z NO: NO:B
X
A
401 3206 pSAC35:CPYSC.d8CK HSAikex2 leader followed by a five pSAC35 1669 1501 1837 2114 2115 HSA/kex2 bl/HSA amino acid linker followed by d8CKbl and mature HSA.
402 3207 pSAC35:GPY.d8CKb1/ HSA/kex2 leader followed by amino acids pSAC35 1670 1502 1838 2116 2117 HSA/kex2 HSA "GPY" followed by d8CKbl and mature HSA.
403 3208 pSAC35:defensin alpha Amino acids A65-C94 of defensin alpha 1 pSAC35 1285 1286 1287 1288 1289 HSA/kex2 (-) 1/HSA fused downstream of the HSA/kex2 leader and upstream of mature HSA.
r; 404 3209 pSAC35:defensin alpha Amino acids C66-C94 of defensin alpha 2 pSAC35 1290 1291 1292 1293 1294 HSA/kex2 (5) 2/HSA fused downstream of the HSA/kex2 leader and upstream of mature HSA.

405 3210 pSAC35:defensin alpha Amino acids 65-94 of SEQ ID N01297, pSAC35 1295 1296 1297 1298 1299 HSA/kex2 3/HSA with A65D and F92I mutations, fused (5) downstream of the HSA/kex2 leader and upstream of mature HSA.
406 3232 pSAC35:CART/HSA HSA/kex2 leader followed by processed pSAC35 1671 1503 1839 2118 2119 HSA/kex2 active cocaine-amphetamine regulated transcript (CART) (amino acids V69 through L116) followed by mature HSA.
407 3238 pSAC35:phosphatonin. Phosphatonin fused upstream of HSA.
pSAC35 1306 1307 1308 Native HSA
phosphaton A
in 408 3270 pSAC35:adipokine/HSA HSA/kex2 leader followed by adipokine pSAC35 1672 1504 1840 2120 2121 HSA/kex2 c7, followed by mature HSA.
oe Table 2 o Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader c...:,...,4 No. ID Vector ID ID ID ID ID Sequence s NO:Y NO: NO:Z NO: NO:B
o 1.., X
A
409 3272 pSAC35.INV: {D} 8CK { CKbeta-1 tandem repeat (x2) fusion to the pSAC35 1673 1505 1841 2122 2123 invertase b}1(x2)/HSA N-termal HSA. Under the invertase signal peptide.
410 3274 pSAC35:Plpal-12.HSA Plpal-12 pepduein peptide fused upstream pSAC35 1334 1335 1336 HSA/kex2 of mature HSA, and downstream of the HSA/kex2 leader sequence.

411 3275 pSAC35:P4pal-10.HSA P4pal-10 pepducin peptide fused upstream pSAC35 1337 1338 1339 HSA/kex2 of mature HSA, and downstream of the I.) a, HSA/kex2 leader sequence.
H
412 3281 pSAC35.PY3- PYY3-36 tandem repeat (x2) fused pSAC35 1674 1506 1842 2124 2125 HSA/kex2 u.) (5) u.) 36(x2)/HSA upstream of HSA and downstream of the I.) HSA/kex2 signal peptide.

a, 413 3282 pSAC35:HSA/PYY3- PYY3-36 tandem repeat (x2) fused pSAC35 1675 1507 1843 2126 2127 HSA/kex2 1 36(x2) downstream of mature HSA and (5) I.) HSA/kex2 leader.
H
414 3298 pSAC35:IL21/HSA Amino acids Q30-S162 of IL-21 fused pSAC35 2167 2157 2177 2188 2189 HSA/Kex2 upstream of mature HSA and downstream of HSA/kex2 leader 415 3307 pSAC35:1L4/HSA Amino acids H25-S153 of IL-4 fiased pSAC35 2168 2158 2178 2190 2191 HSA/Kex2 upstream of mature HSA and downstream of HSA/kex2 leader 416 3309 pSAC:KT.GLP-1(7- Killer toxin leader sequence followed by pSAC351-d 2170 2160 2180 2194 2195 Killer toxin n 36(A8G))x2.MSA.E25- GLP-1(7-36(A8G) followed by mature A608 mouse serum albumin.
cp o 417 3312 pSAC35:h0CIL/HSA HSA/kex2 leader followed by amino pSAC35 2171 2161 2181 2196 2197 HSA/Kex2 4-t--;
acids N20 to V149 of h0CIL followed by o oe o mature HSA
1¨, =
Table 2 Fusion Construct Construct Name Description Expression SEQ SEQ SEQ SEQ SEQ Leader No. ID Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
418 7777 T20:HSA T20 fused downstream of full length HSA
pC4 1170 1171 1172 HSA
419 8888 pC4:BNP.HSA Human B-type natriuretic peptide fused pC4 1275 1276 1277 1278 1279 Native upstream of mature HSA.
BNP
420 9999 T1249:HSA T1249 fused downstream of full length pC4 1173 1174 1175 HSA
HSA

=
t=.) =

[0066] Table 2 provides a non-exhaustive list of polynucleotides of the invention comprising, or alternatively consisting of, nucleic acid molecules encoding an albumin fusion protein. The first column, "Fusion No." gives a fusion number to each polynucleotide.
Column 2, "Construct ID" provides a unique numerical identifier for each polynucleotide of the invention. The Construct IDs may be used to identify polynucleotides which encode albumin fusion proteins comprising, or alternatively consisting of, a Therapeutic protein portion corresponding to a given Therapeutic Protein:X listed in the corresponding row of Table 1 wherein that Construct ID is listed in column 5. The "Construct Name"
column (column 3) provides the name of a given albumin fusion construct or polynucleotide.
[0067] The fourth column in Table 2, "Description" provides a general description of a given albumin fusion construct, and the fifth column, "Expression Vector"
lists the vector into which a polynucleotide comprising, or alternatively consisting of, a nucleic acid molecule encoding a given albumin fusion protein was cloned. Vectors are known in the art, and are available commercially or described elsewhere. For example, as described in the Examples, an "expression cassette" comprising, or alternatively consisting of, one or more of (1) a polynucleotide encoding a given albumin fusion protein, (2) a leader sequence, (3) a promoter region, and (4) a transcriptional terminator, may be assembled in a convenient cloning vector and subsequently be moved into an alternative vector, such as, for example, an expression vector including, for example, a yeast expression vector or a mammalian expression vector. In one embodiment, for expression in S. cervisiae, an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pSAC35. In another embodiment, for expression in CHO cells, an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pC4. In a further embodiment, a polynucleotide comprising or alternatively consisting of a nucleic acid molecule encoding the Therapeutic protein portion of an albumin fusion protein is cloned into pC4:HSA. In a still further embodiment, for expression in NSO cells, an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pEE12. Other useful cloning and/or expression vectors will be known to the skilled artisan and are within the scope of the invention.
[0068] Column 6, "SEQ ID NO:Y," provides the full length amino acid sequence of the albumin fusion protein of the invention, hr most instances, SEQ ID NO:Y shows the unprocessed form of the albumin fusion protein encoded ¨ in other words, SEQ
ID NO:Y

shows the signal sequence, a HSA portion, and a therapeutic portion all encoded by the particular construct.
Specifically contemplated by the present invention are all polynucleotides that encode SEQ ID NO:Y. When these polynucleotides are used to express the encoded protein from a cell, the cell's natural secretion and processing steps produces a protein that lacks the signal sequence listed in columns 4 and/or 11 of Table 2. The specific amino acid sequence of the listed signal sequence is shown later in the specification or is well known in the art. Thus, most preferred embodiments of the present invention include the albumin fusion protein produced by a cell (which would lack the leader sequence shown in columns 4 and/or 11 of Table 2). Also most preferred are polypeptides comprising SEQ ID
NO:Y without the specific leader sequence listed in columns 4 and/or 11 of Table 2.
Compositions comprising these two preferred embodiments, including pharmaceutical compositions, are also preferred. Moreover, it is well within the ability of the skilled artisan to replace the signal sequence listed in columns 4 and/or 11 of Table 2 with a different signal sequence, such as those described later in the specification to facilitate secretion of the processed albumin fusion protein.
[0069] The seventh column, "SEQ ID NO:X," provides the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of a given albumin , fusion protein may be derived. In one embodiment, the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein may be derived comprises the wild type gene sequence encoding a Therapeutic protein shown in Table 1. In an alternative embodiment, the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein may be derived comprises a variant or derivative of a wild type gene sequence encoding a Therapeutic protein shown in Table 1, such as, for example, a synthetic codon optimized variant of a wild type gene sequence encoding a Therapeutic protein.
[0070] The eighth column, "SEQ ID NO:Z," provides a predicted translation of the parent nucleic acid sequence (SEQ ID NO:X). This parent sequence can be a full length parent protein used to derive the particular construct, the mature portion of a parent protein, a variant or fragment of a wildtype protein, or an artificial sequence that can be used to create the described construct. One of skill in the art can use this amino acid sequence shown in SEQ
ID NO:Z to determine which amino acid residues of an albumin fusion protein encoded by a given construct are provided by the therapeutic protein. Moreover, it is well within the ability of the skilled artisan to use the sequence shown as SEQ ID NO:Z to derive the construct described in the same row. For example, if SEQ ID NO:Z corresponds to a full length protein, but only a portion of that protein is used to generate the specific CID, it is within the skill of the art to rely on molecular biology techniques, such as PCR, to amplify the specific fragment and clone it into the appropriate vector.
[0071] Amplification primers provided in columns 9 and 10, "SEQ ID NO:A"
and "SEQ
ID NO:B" respectively, are exemplary primers used to generate a polynucleotide comprising or alternatively consisting of a nucleic acid molecule encoding the Therapeutic protein portion of a given albumin fusion protein. In one embodiment of the invention, oligonucleotide primers having the sequences shown in columns 9 and/or 10 (SEQ
ID NOS:A
and/or B) are used to PCR amplify a polynucleotide encoding the Therapeutic protein portion of an albumin fusion protein using a nucleic acid molecule comprising or alternatively consisting of the nucleotide sequence provided in column 7 (SEQ ID NO:X)of the corresponding row as the template DNA. PCR methods are well-established in the art.
Additional useful primer sequences could readily be envisioned and utilized by those of ordinary skill in the art.
[0072] In an alternative embodiment, oligonucleotide primers may be used in overlapping PCR reactions to generate mutations within a template DNA sequence. PCR
methods are known in the art.
[0073] As shown in Table 3, certain albumin fusion constructs disclosed in this application have been deposited with the ATCCS.

Table 3 Construct ID Construct Name ATCC Deposit No./ Date 1642 pSAC35:GCSF.T31-P204.HSA PTA-3767 Oct. 5, 2001 1643 pSAC35:HSA.GCSF.T31-P204 PTA-3766 Oct. 5, 2001 1812 pSAC35:IL2.A21-T153.HSA PTA-3759 Oct. 4, 2001 1941 pC4:HSA/PTH84(junctioned) PTA-3761 Oct. 4, 2001 1949 pC4:PTH.S1-Q84/HSA (junctioned) PTA-3762 Oct. 4, 2001 1966 pC4:EPO.M1-D192.HSA PTA-3771 Oct. 5, 2001 also named pC4:EPOM1-D192.HSA
1981 pC4.HSA-EPO.A28-D192 PTA-3770 Oct. 5, 2001 1997 pEE12.1:EPOM1-D192.HSA PTA-3768 Oct. 5, 2001 2030 pSAC35.ycolL-2.A21-T153.HSA PTA-3757 Oct. 4, 2001 2031 pSAC35.HSA.ycoIL-2.A21-T153 PTA-3758 Oct. 4, 2001 2053 pEE12:1FNb-HSA PTA-3764 Oct. 4, 2001 also named pEE12.1:IFNI3-HSA
2054 pEE12:HSA-IFNb PTA-3941 Dec. 19, 2001 2249 pSAC35:IFNa2-HSA PTA-3763 Oct. 4, 2001 also named pSAC23:IFNa2-HSA
2250 pSAC35:HSAINSULIN(GYG) PTA-3916 Dec. 07, 2001 also named pSAC35.HSA.INSULING(GYG).F1-N62 2255 pSAC35:INSULIN(GYG).HSA PTA-3917 Dec. 07, 2001 also named pSAC35.INSULING(GYG).F1-N62.HSA
2276 pSAC35:HSA.INSULIN(GGG) PTA-3918 Dec. 07, 2001 also named pSAC35.HSA.INSULING(GGG) .F1-N58 2298 pEE12.1:EPO.R140G.HSA PTA-3760 Oct. 4, 2001 Construct ID Construct Name ATCC Deposit No./ Date 2294 pC4:EPO.R140G.HSA PTA-3742 Sept. 28, 2001 also named pC4.EPO.R1406.HSA
2325 pC4.EPO:Ml-D192.HSA.Codon opt. PTA-3773 Oct. 5, 2001 2343 pSAC35.lNV-IFNA2.HSA PTA-3940 Dec. 19, 2001 2363 pC4.GCSF.HSA.EPO.A28-D192 PTA-3740 Sept. 28, 2001 2373 pC4.GCSF.HSA.EPO.A28-D192.R140G PTA-3741 Sept. 28, 2001 2381 pC4:HSA-IFNa2(C17-E181) PTA-3942 Dec. 19, 2001 2382 pC4:IFNa2-HSA PTA-3939 Dec. 19, 2001 2387 pC4:EPO(G140)-HSA-GCSF.T31-P204 PTA-3919 Dec. 11, 2001 2414 pC4.EPO:M1-D192copt.HSA.GCSF.T31-P204 PTA-3924 Dec. 12, 2001 also named pC4.EPO:M1-D192copt.HAS.GCSF.T31-P204 2441 pEE12.EPO:Ml-D192copt.HSA.GCSF.T31- PTA-3923 P204 Dec. 12, 2001 also named:
pEE12.EPO:M1-D192copt.HAS.GCSF.T31-2492 pC4.1ENb(deltaM22).HSA PTA-3943 Dec. 19, 2001 3070 pSAC35:KT.GLP-1(7-36(A8G))x2.HSA PTA-4671 Sept. 16, 2002 3165 pSAC35:HSA.IFNa PTA-4670 Sept. 16, 2002 also named CID 3165, pSAC35:HSA.INFa 3163 pSAC35:HSA.hGH PTA-4770 October 22, 2002 [0074] It is possible to retrieve a given albumin fusion construct from the deposit by techniques known in the art and described elsewhere herein (see, Example 40).
The ATCC is located at 10801 University Boulevard, Manassas, Virginia 20110-2209, USA. The ATCC
deposits were made pursuant to the terms of the Budapest Treaty on the international recognition of the deposit of microorganisms for the purposes of patent procedure.

[0075] In a further embodiment of the invention, an "expression cassette" comprising, or alternatively consisting of one or more of (1) a polynucleotide encoding a given albumin fusion protein, (2) a leader sequence, (3) a promoter region, and (4) a transcriptional terminator can be moved or "subcloned" from one vector into another. Fragments to be subcloned may be generated by methods well known in the art, such as, for example, PCR
amplification (e.g., using oligonucleotide primers having the sequence shown in SEQ ID
NO:A or B), and/or restriction enzyme digestion.
[0076] In preferred embodiments, the albumin fusion proteins of the invention are capable of a therapeutic activity and/or biologic activity corresponding to the therapeutic activity and/or biologic activity of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein listed in the corresponding row of Table 1. In further preferred embodiments, the therapeutically active protein portions of the albumin fusion proteins of the invention are fragments or variants of the protein encoded by the sequence shown in SEQ ID NO:X column of Table 2, and are capable of the therapeutic activity and/or biologic activity of the corresponding Therapeutic protein.
Non-human albumin fusion proteins of growth hormone.
[0077] In one embodiment, the albumin fusion proteins of the invention comprise one or more Serum Albumin proteins of a non-human animal species, fused in tandem and in-frame either at the N-teiminus or the C-terminus to one or more Growth Hormone proteins of the same non-human animal species. Non-human Serum Albumin and Growth Hormone proteins are well known in the art and available in public databases. For example, Table 4 presents accession numbers corresponding to non-human Serum Albumin sequences (column 2) and non-human Growth Hormone sequences (column 3) found in GenBank. In a preferred embodiment, a Serum Albumin protein from a non-human animal species listed in Table 4 is fused to a Growth Hormone protein from the same non-human animal species.
[0078] In a specific embodiment, the albumin fusion protein of the invention comprises one or more Bos taurus Serum Albumin proteins listed in Table 4, column 2, fused in tandem and in-frame either at the N-terminus or the C-terminus to one or more Bos taurus Growth Hormone proteins listed in Table 4, column 3.
[0079]
Fusion proteins comprising fragments or variants of non-human Serum Albumin, such as, for example, the mature form of Serum Albumin, are also encompassed by the invention. Fusion proteins comprising fragments or variants of non-human Growth Hormone proteins, such as, for example, the mature form of Growth Hormone, are also encompassed by the invention. Preferably the non-human Growth Hormone fragments and variants retain growth hormone activity.
[0080] Polynucleotides of the invention comprise, or alternatively consist of, one or more nucleic acid molecules encoding a non-human albumin fusion protein described above.
For example, the polynucleotides can comprise, or alternatively consist of, one or more nucleic acid molecules that encode a Serum Albumin protein from a non-human animal species listed in Table 4, column 1 (such as, for example, the non-human Serum Albumin reference sequences listed in Table 4, column 2) fused in tandem and in-frame either 5' or 3' to a polynucleotide that comprises, or alternatively consists of, one or more nucleic acid molecules encoding the non-human Growth Hormone protein of the corresponding non-human animal species (for example, the Growth Hormone reference sequences listed in Table 4, column 3).
[0081] The above-described non-human albumin fusion proteins are encompassed by the invention, as are host cells and vectors containing these polynucleotides.
In one embodiment, a non-human albumin fusion protein encoded by a polynucleotide as described above has extended shelf life. In an additional embodiment, a non-human albumin fusion protein encoded by a polynucleotide described above has a longer serum half-life and/or more stabilized activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo than the corresponding unfused Growth Hormone molecule.
[0082] The present invention also encompasses methods of preventing, treating, or ameliorating a disease or disorder in a non-human animal species. In certain embodiments, the present invention encompasses a method of treating a veterinary disease or disorder comprising administering to a non-human animal species in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Growth Hormone portion corresponding to a Growth Hormone protein (or fragment or variant thereof) in an amount effective to treat, prevent or ameliorate the disease or disorder.
Veterinary diseases and/or disorders which may be treated, prevented, or ameliorated include growth disorders (such as, for example, pituitary dwarfism), shin soreness, obesity, growth hormone-responsive dermatosis, dilated cardiomyopathy, eating disorders, reproductive disorders, and endocrine disorders.
[0083] Non-human albumin fusion proteins of the invention may also be used to promote healing of skin wounds, corneal injuries, bone fractures, and injuries of joints, tendons, or ligaments.
[0084] Non-human albumin fusion proteins of the invention may also be used to increase milk production in lactating animals. In a preferred embodiment, the lactating animal is a dairy cow.
[0085] Non-human albumin fusion proteins of the invention may also be used to improve body condition in aged animals.
[0086] Non-human albumin fusion proteins of the invention may also be used to increase fertility, pregnancy rates, and reproductive success in domesticated animals.
[0087] Non-human albumin fusion proteins of the invention may also be used to improve the lean-to-fat ratio in animals raised for consumption, as well as to improve appetite, and increase body size and growth rate.
Table 4 Non-Human Non-Human Serum Albumin Non-Human Growth Hormone Species Reference Sequence(s):
Reference Sequence(s): GenBank GenBank Protein Accession Protein Accession Nos.
Nos.
Bos taurus ABBOS, CAA76847, P02769, STBO, BAA06379, A29864, CAA41735, 229552, AAF28806, AAF28805, AAF'28804, AAA51411 P01246, AAF03132, AAC63901, AAB92549, A36506,145901, JC1316, CAA23445, CAA00787, CAA00598, AAA30547, AAA30546, AAA30545, AAA30544, AAA30543, AAA30542 Sus scrofa P08835, CAA30970, STPG, PC1017, AAB29947, AAA30988 AAB84359, 146585,146584, PC1063, A01516, AAB17619, 226829, 225740, CAA37411, CAA00592, AAA73478, AAA73477, CAA00356, AAA31046, AAA31045, AAA31044, AA30543 Equus caballus ABHOS, AAG40944, P35747, STHO, P01245, AAD25992, 227704, Ovis aries ABSHS, P14639, CAA34903 STSH, AAB24467, AAC48679, 228487, 223932, CAA34098, CAA31063, CAA00828, AAA31527 Salm salar ABONS2, ABONS2, STONC, P07064, Q07221, P48096, CAA36643, CAA43187 P10814, P10607, 151186, S03709, JS0179, A23154, S06489, CAA42431, AAB29165, AAB24612, Q91221, Q91222, CAA43942, CAA32481, 738042, 224555, CAA00427, AAA50757, AAA49558, AAA49555, Non-Human Non-Human Serum Albumin Non-Human Growth Hormone Species Reference Sequence(s):
Reference Sequence(s): GenBank GenBank Protein Accession Protein Accession Nos.
Nos.
AAA49553, AAA49401, AAA49406, AAA49403, AAA49402 Gallus gallus ABCHS, P19121, CAA43098 BAB62262, BAB69037, AAK95643, A60509, AAG01029, BAA01365, P08998, 226895, CAA31127, CAA35619, AAA48780 Felis catus P49064, S57632, CAA59279, JC4632, P46404, AAC00073, JC4660 AAA96142, AAA67294 Canis P49822, S29749, CAB64867, P33711, 146145, AA1F89582, familiaris CAA76841, AAB30434 AAF21502, AAD43366, S35790, AAB34229, CAA80601 Polypeptide and Polynucleotide Fragments and Variants Fragments [0088] The present invention is further directed to fragments of the Therapeutic proteins described in Table 1, albumin proteins, and/or albumin fusion proteins of the invention.
[0089] The present invention is also directed to polynucleotides encoding fragments of the Therapeutic proteins described in Table 1, albumin proteins, and/or albumin fusion proteins of the invention.
[0090] Even if deletion of one or more amino acids from the N-terminus of a protein results in modification or loss of one or more biological functions of the Therapeutic protein, albumin protein, and/or albumin fusion protein of the invention, other Therapeutic activities and/or functional activities (e.g., biological activities, ability to multimerize, ability to bind a ligand) may still be retained. For example, the ability of polypeptides with N-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptides generally will be retained when less than the majority of the residues of the complete polypeptide are removed from the N-terminus. Whether a particular polypeptide lacking N-terminal residues of a complete polypeptide retains such immunologic activities can readily be determined by routine methods described herein and otherwise known in the art. It is not unlikely that a mutein with a large number of deleted N-terminal amino acid residues may retain some biological or immunogenic activities. In fact, peptides composed of as few as six amino acid residues may often evoke an immune response.
[0091] Accordingly, fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2). In particular, N-terminal deletions may be described by the general formula m to q, where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2), and m is defined as any integer ranging from 2 to q minus 6. Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0092] In addition, fragments of serum albumin polypeptides corresponding to an albumin protein portion of an albumin fusion protein of the invention, include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., serum albumin, or a serum albumin portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2). In preferred embodiments, N-terminal deletions may be described by the general formula m to 585, where 585 is a whole integer representing the total number of amino acid residues in mature human serum albumin (SEQ ID NO:1038), and m is defined as any integer ranging from 2 to 579. Polynucleotides encoding these polypeptides are also encompassed by the invention. In additional embodiments, N-terminal deletions may be described by the general formula m to 609, where 609 is a whole integer representing the total number of amino acid residues in full length human serum albumin (SEQ ID
NO:1094), and m is defined as any integer ranging from 2 to 603. Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0093] Moreover, fragments of albumin fusion proteins of the invention, include the full length albumin fusion protein as well as polypeptides having one or more residues deleted from the amino terminus of the albumin fusion protein (e.g., an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2; or an albumin fusion protein having the amino acid sequence disclosed in column 6 of Table 2). In particular, N-terminal deletions may be described by the general formula m to q, where q is a whole integer representing the total number of amino acid residues in the albumin fusion protein, and m is defined as any integer ranging from 2 to q minus 6.
Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0094] Also as mentioned above, even if deletion of one or more amino acids from the N-terminus or C-terminus of a reference polypeptide (e.g., a Therapeutic protein; serum albumin protein; or albumin fusion protein of the invention) results in modification or loss of one or more biological functions of the protein, other functional activities (e.g., biological activities, ability to multimerize, ability to bind a ligand) and/or Therapeutic activities may still be retained. For example the ability of polypeptides with C-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptide generally will be retained when less than the majority of the residues of the complete or mature polypeptide are removed from the C-terminus. Whether a particular polypeptide lacking the N-terminal and/or C-terminal residues of a reference polypeptide retains Therapeutic activity can readily be determined by routine methods described herein and/or otherwise known in the art.
[0095] The present invention further provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2). In particular, C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to q minus 1, and where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2). Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0096] In addition, the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention (e.g., serum albumin or an albumin protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2). In particular, C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to 584, where 584 is the whole integer representing the total number of amino acid residues in mature human serum albumin (SEQ ID NO:1038) minus 1.
Polynucleotides encoding these polypeptides are also encompassed by the invention. In particular, C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to 608, where 608 is the whole integer representing the total number of amino acid residues in serum albumin (SEQ ID NO:1094) minus 1. Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0097] Moreover, the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of an albumin fusion protein of the invention. In particular, C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to q minus 1, and where q is a whole integer representing the total number of amino acid residues in an albumin fusion protein of the invention.
Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0098] In addition, any of the above described N- or C-terminal deletions can be combined to produce a N- and C-tenninal deleted reference polypeptide. The invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini, which may be described generally as having residues m to n of a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or serum albumin (e.g., SEQ ID NO:1038), or an albumin protein portion of an albumin fusion protein of the invention, or an albumin protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or an albumin fusion protein, or an albumin fusion protein encoded by a polynucleotide or albumin fusion construct of the invention) where n and m are integers as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0099] The present application is also directed to proteins containing polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a reference polypeptide sequence (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or serum albumin (e.g., SEQ
ID NO: 1038), or an albumin protein portion of an albumin fusion protein of the invention, or an albumin protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or an albumin fusion protein, or an albumin fusion protein encoded by a polynucleotide or albumin fusion construct of the invention) set forth herein, or fragments thereof. In preferred embodiments, the application is directed to proteins comprising polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to reference polypeptides having the amino acid sequence of N- and C-terminal deletions as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0100] Preferred polypeptide fragments of the invention are fragments comprising, or alternatively, consisting of, an amino acid sequence that displays a Therapeutic activity and/or functional activity (e.g. biological activity) of the polypeptide sequence of the Therapeutic protein or serum albumin protein of which the amino acid sequence is a fragment.
[0101] Other preferred polypeptide fragments are biologically active fragments.
Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide of the present invention. The biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity.
Variants [0102] "Variant" refers to a polynucleotide or nucleic acid differing from a reference nucleic acid or polypeptide, but retaining essential properties thereof Generally, variants are overall closely similar, and, in many regions, identical to the reference nucleic acid or polypeptide.
[0103] As used herein, "variant", refers to a Therapeutic protein portion of an albumin fusion protein of the invention, albumin portion of an albumin fusion protein of the invention, or albumin fusion protein of the invention differing in sequence from a Therapeutic protein (e.g. see "therapeutic" column of Table 1), albumin protein, and/or albumin fusion protein, respectively, but retaining at least one functional and/or therapeutic property thereof as described elsewhere herein or otherwise known in the art. Generally, variants are overall very similar, and, in many regions, identical to the amino acid sequence of the Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein, albumin protein corresponding to an albumin protein portion of an albumin fusion protein, and/or albumin fusion protein. Nucleic acids encoding these variants are also encompassed by the invention.
[0104] The present invention is also directed to proteins which comprise, or alternatively consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, identical to, for example, the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., the amino acid sequence of a Therapeutic protein:X disclosed in Table 1; or the amino acid sequence of a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 1 and 2, or fragments or variants thereof), albumin proteins corresponding to an albumin protein portion of an albumin fusion protein of the invention (e.g., the amino acid sequence of an albumin protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 1 and 2; the amino acid sequence shown in SEQ ID
NO: 1038;
or fragments or variants thereof), and/or albumin fusion proteins. Fragments of these polypeptides are also provided (e.g., those fragments described herein).
Further polypeptides encompassed by the invention are polypeptides encoded by polynucleotides which hybridize to the complement of a nucleic acid molecule encoding an albumin fusion protein of the invention under stringent hybridization conditions (e.g., hybridization to filter bound DNA in 6X Sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.2X SSC, 0.1% SDS at about 50 - 65 degrees Celsius), under highly stringent conditions (e.g., hybridization to filter bound DNA in 6X sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.1X SSC, 0.2% SDS at about 68 degrees Celsius), or under other stringent hybridization conditions which are known to those of skill in the art (see, for example, Ausubel, F.M.
et al., eds., 1989 Current protocol in Molecular Biology, Green publishing associates, Inc., and John Wiley &
Sons Inc., New York, at pages 6.3.1 - 6.3.6 and 2.10.3). Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0105] By a polypeptide having an amino acid sequence at least, for example, 95%
"identical" to a query amino acid sequence, it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a query amino acid sequence, up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, or substituted with another amino acid. These alterations of the reference sequence may occur at the amino- or carboxy-terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
[0106] As a practical matter, whether any particular polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid sequence of an albumin fusion protein of the invention or a fragment thereof (such as a Therapeutic protein portion of the albumin fusion protein or an albumin portion of the albumin fusion protein), can be determined conventionally using known computer programs. A preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al.
(Comp. App. Biosci.6:237-245 (1990)). In a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences.
The result of said global sequence alignment is expressed as percent identity. Preferred parameters used in a FASTDB amino acid alignment are: Matrix=PAM 0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=1, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter.
[0107] If the subject sequence is shorter than the query sequence due to N- or C-terminal deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for N- and C-teitninal truncations of the subject sequence when calculating global percent identity.
For subject sequences truncated at the N- and C-tetwini, relative to the query sequence, the percent identity is corrected by calculating the number of residues of the query sequence that are N-and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence.
Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment.
This percentage is then subtracted from the percent identity, calculated by the above FASTDB
program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence.
[0108] For example, a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity. The deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus. The 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%. In another example, a 90 residue subject sequence is compared with a 100 residue query sequence. This time the deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends- of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
[0109] The variant will usually have at least 75 % (preferably at least about 80%, 90%, 95% or 99%) sequence identity with a length of normal HA or Therapeutic protein which is the same length as the variant. Homology or identity at the nucleotide or amino acid sequence level is determined by BLAST (Basic Local Alignment Search Tool) analysis using the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx (Karlin et al., Proc. Natl. Acad. Sci. USA .87: 2264-2268 (1990) and Altschul, J. Mol.
Evol. 36:
290-300 (1993)) which are tailored for sequence similarity searching.
[0110] The approach used by the BLAST program is to first consider similar segments between a query sequence and a database sequence, then to evaluate the statistical significance of all matches that are identified and finally to summarize only those matches which satisfy a preselected threshold of significance. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al., (Nature Genetics 6: 119-129 (1994)), The search parameters for histogram, descriptions, alignments, expect (i.e., the statistical significance threshold for reporting matches against database sequences), cutoff, matrix and filter are at the default settings. The default scoring matrix used by blastp, blastx, tblastn, and tblastx is the BLOSUM62 matrix (Henikoff et al., Proc. Natl. Acad. Sci. USA 89: 10915-10919 (1992).
For blastn, the scoring matrix is set by the ratios of M (i.e., the reward score for a pair of matching residues) to N (i.e., the penalty score for mismatching residues), wherein the default values for M and N are 5 and -4, respectively. Four blastn parameters may be adjusted as follows: Q=10 (gap creation penalty); R=10 (gap extension penalty); wink=1 (generates word hits at every winkth position along the query); and gapw=16 (sets the window width within which gapped alignments are generated). The equivalent Blastp parameter settings were Q=9; R=2; wink=1; and gapw=32. A Bestfit comparison between sequences, available in the GCG package version 10.0, uses DNA parameters GAP=50 (gap creation penalty) and LEN=3 (gap extension penalty) and the equivalent settings in protein comparisons are GAP=8 and LEN=2.
[0111] The polynucleotide variants of the invention may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred.
Moreover, polypeptide variants in which less than 50, less than 40, less than 30, less than 20, less than 10, or 5-50, 5-25, 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are also preferred. Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host, such as, yeast or E. coli).
[0112] In a preferred embodiment, a polynucleotide of the invention which encodes the albumin portion of an albumin fusion protein is optimized for expression in yeast or mammalian cells. In a further preferred embodiment, a polynucleotide of the invention which encodes the Therapeutic protein portion of an albumin fusion protein is optimized for expression in yeast or mammalian cells. In a still further preferred embodiment, a polynucleotide encoding an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
[0113] In an alternative embodiment, a codon optimized polynucleotide which encodes a Therapeutic protein portion of an albumin fusion protein does not hybridize to the wild type polynucleotide encoding the Therapeutic protein under stringent hybridization conditions as described herein. In a further embodiment, a codon optimized polynucleotide which encodes an albumin portion of an albumin fusion protein does not hybridize to the wild type polynucleotide encoding the albumin protein under stringent hybridization conditions as described herein. In another embodiment, a codon optimized polynucleotide which encodes an albumin fusion protein does not hybridize to the wild type polynucleotide encoding the Therapeutic protein portion or the albumin protein portion under stringent hybridization conditions as described herein.
[0114] In an additional embodiment, a polynucleotide which encodes a Therapeutic protein portion of an albumin fusion protein does not comprise, or alternatively consist of, the naturally occurring sequence of that Therapeutic protein. In a further embodiment, a polynucleotide which encodes an albumin protein portion of an albumin fusion protein does not comprise, or alternatively consist of, the naturally occurring sequence of albumin protein.
In an alternative embodiment, a polynucleotide which encodes an albumin fusion protein does not comprise, or alternatively consist of, the naturally occurring sequence of a Therapeutic protein portion or the albumin protein portion.
[0115] Naturally occurring variants are called "allelic variants," and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism.
(Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)). These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
[0116] Using known methods of protein engineering and recombinant DNA
technology, variants may be generated to improve or alter the characteristics of the polypeptides of the present invention. For instance, one or more amino acids can be deleted from the N-terminus or C-terminus of the polypeptide of the present invention without substantial loss of biological function. As an example, Ron et al. (J. Biol.
Chem. 268: 2984-2988 (1993)) reported variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-216 (1988).) [0117] Moreover, ample evidence demonstrates that variants often retain a biological activity similar to that of the naturally occurring protein. For example, Gayle and coworkers (J. Biol. Chem. 268:22105-22111(1993)) conducted extensive mutational analysis of human cytokine IL-la. They used random mutagenesis to generate over 3,500 individual IL-la mutants that averaged 2.5 amino acid changes per variant over the entire length of the molecule. Multiple mutations were examined at every possible amino acid position. The investigators found that "[m]ost of the molecule could be altered with little effect on either [binding or biological activity]." In fact, only 23 unique amino acid sequences, out of more than 3,500 nucleotide sequences examined, produced a protein that significantly differed in activity from wild-type.
[0118] Furthermore, even if deleting one or more amino acids from the N-terminus or C-terminus of a polypeptide results in modification or loss of one or more biological functions, other biological activities may still be retained. For example, the ability of a deletion variant to induce and/or to bind antibodies which recognize the secreted form will likely be retained when less than the majority of the residues of the secreted form are removed from the N-terminus or C-terminus. Whether a particular polypeptide lacking N- or C-terminal residues of a protein retains such immunogenic activities can readily be determined by routine methods described herein and otherwise known in the art.
[0119] Thus, the invention further includes polypeptide variants which have a functional activity (e.g., biological activity and/or therapeutic activity).
In one embodiment, the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity) that corresponds to one or more biological and/or therapeutic activities of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein. In another embodiment, the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity) that corresponds to one or more biological and/or therapeutic activities of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein. Such variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity. Polynucleotides encoding such variants are also encompassed by the invention.
[0120] In preferred embodiments, the variants of the invention have conservative substitutions. By "conservative substitutions" is intended swaps within groups such as replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile;
replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu;
replacement of the amide residues Asn and Gin, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly.
[0121] Guidance concerning how to make phenotypically silent amino acid substitutions is provided, for example, in Bowie et al., "Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990), wherein the authors indicate that there are two main strategies for studying the tolerance of an amino acid sequence to change.
[0122] The first strategy exploits the tolerance of amino acid substitutions by natural selection during the process of evolution. By comparing amino acid sequences in different species, conserved amino acids can be identified. These conserved amino acids are likely important for protein function. In contrast, the amino acid positions where substitutions have been tolerated by natural selection indicates that these positions are not critical for protein function. Thus, positions tolerating amino acid substitution could be modified while still maintaining biological activity of the protein.
[0123] The second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critical for protein function. For example, site directed mutagenesis or alanine-scanning mutagenesis (introduction of single alanine mutations at every residue in the molecule) can be used. See Cunningham and Wells, Science 244:1081-1085 (1989). The resulting mutant molecules can then be tested for biological activity.
[0124] As the authors state, these two strategies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors further indicate which amino acid changes are likely to be permissive at certain amino acid positions in the protein. For example, most buried (within the tertiary structure of the protein) amino acid residues require nonpolar side chains, whereas few features of surface side chains are generally conserved.
Moreover, tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu;
replacement of the amide residues Asn and Gln, replacement of the basic residues Lys, Arg, and His;
replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly. Besides conservative amino acid substitution, variants of the present invention include (i) polypeptides containing substitutions of one or more of the non-conserved amino acid residues, where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) polypeptides containing substitutions of one or more of the amino acid residues having a substituent group, or (iii) polypeptides which have been fused with or chemically conjugated to another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), (iv) polypeptide containing additional amino acids, such as, for example, an IgG Fc fusion region peptide . Such variant polypeptides are deemed to be within the scope of those skilled in the art from the teachings herein.
[0125]
For example, polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as less aggregation.
Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity. See Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993).
[0126] In specific embodiments, the polypeptides of the invention comprise, or alternatively, consist of, fragments or variants of the amino acid sequence of an albumin fusion protein, the amino acid sequence of a Therapeutic protein and/or human serum albumin, wherein the fragments or variants have 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, amino acid residue additions, substitutions, and/or deletions when compared to the reference amino acid sequence. In preferred embodiments, the amino acid substitutions are conservative. Nucleic acids encoding these polypeptides are also encompassed by the invention.
[0127]
The polypeptide of the present invention can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids. The polypeptides may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of fiavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
(See, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993); POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth. Enzymol.
182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48-62 (1992)).
Functional activity [0128] "A polypeptide having functional activity" refers to a polypeptide capable of displaying one or more known functional activities associated with the full-length, pro-protein, and/or mature form of a Therapeutic protein. Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti-polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polyp eptide .
[0129] "A polypeptide having biological activity" refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a Therapeutic protein of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide of the present invention (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention).
[0130] In preferred embodiments, an albumin fusion protein of the invention has at least one biological and/or therapeutic activity associated with the Therapeutic protein portion (or fragment or variant thereof) when it is not fused to albumin.

[0131] The albumin fusion proteins of the invention can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein. Additionally, one of skill in the art may routinely assay fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein, for activity using assays referenced in its corresponding row of Table 1 (e.g., in column 3 of Table 1). Further, one of skill in the art may routinely assay fragments of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein, for activity using assays known in the art and/or as described in the Examples section below.
[0132] For example, in one embodiment where one is assaying for the ability of an albumin fusion protein to bind or compete with a Therapeutic protein for binding to an anti-Therapeutic polypeptide antibody and/or anti-albumin antibody, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc. In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
[0133] In a preferred embodiment, where a binding partner (e.g., a receptor or a ligand) of a Therapeutic protein is identified, binding to that binding partner by an albumin fusion protein which comprises that Therapeutic protein as the Therapeutic protein portion of the fusion can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995). In another embodiment, the ability of physiological correlates of an albumin fusion protein to bind to a substrate(s) of the Therapeutic polypeptide corresponding to the Therapeutic protein portion of the fusion can be routinely assayed using techniques known in the art.
[0134] In an alternative embodiment, where the ability of an albumin fusion protein to roultimerize is being evaluated, association with other components of the multimer can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., supra.
[0135] In preferred embodiments, an albumin fusion protein comprising all or a portion of an antibody that binds a Therapeutic protein, has at least one biological and/or therapeutic activity (e.g., to specifically bind a polypeptide or epitope) associated with the antibody that binds a Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin. In other preferred embodiments, the biological activity and/or therapeutic activity of an albumin fusion protein comprising all or a portion of an antibody that binds a Therapeutic protein is the inhibition (i.e., antagonism) or activation (i.e., agonism) of one or more of the biological activities and/or therapeutic activities associated with the polyp eptide that is specifically bound by antibody that binds a Therapeutic protein.
[0136] Albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be characterized in a variety of ways. In particular, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art.
[0137] Assays for the ability of the albumin fusion proteins (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Patent No. 5,223,409), on spores (e.g., Patent Nos. 5,571,698; 5,403,484; and 5,223,409), on plastnids (e.g., Cull et al., Proc. Natl.
Acad. Sci. USA 89:1865-1869 (1992)) or on phage (e.g., Scott and Smith, Science 249:386-390 (1990); Devlin, Science 249:404-406 (1990); Cwirla et al., Proc.
Natl. Acad.
Sci. USA 87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)).
Albumin fusion proteins comprising at least a fragment or variant of a Therapeutic antibody may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art.

[0138] The albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art.
[0139]
Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA
(enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York).
Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
[0140]
Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as 111PA buffer (1% NP-40 or Tritoli X-100, 1% sodium deoxycholate, TM
0.1% SDS, 0.15 M NaC1, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinM, sodium .vanadate), adding the albumin fusion protein of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C, adding sepharosTem beads coupled to an anti-albumin antibody, for example, to the cell lysate, incubating for about an hour or more at 40 degrees C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the albumin fusion protein to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sephartabeads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.

f0141] Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20%
SDS-PAGE
depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylainide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tweer 20), applying the albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 125I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
[0142] ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the albumin fusion protein (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the albumin fusion proteins specifically bound to the antigen coating the well. In ELISAs the albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes albumin fusion protein) conjugated to a detectable compound may be added to the well.
Further, instead of coating the well with the antigen, the albumin fusion protein may be coated to the well. In this case, the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase). One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.
[0143] The binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding assays. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 1251) with the albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen. The affinity of the albumin fusion protein for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis. Competition with a second protein that binds the same protein, antigen or epitope as the albumin fusion protein, can also be determined using radioimmunoassays. In this case, the protein, antigen or epitope is incubated with an albumin fusion protein conjugated to a labeled compound (e.g., 3H or 1251) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
101441 In a preferred embodiment, BlAcore kinetic analysis is used to determine the binding on and off rates of albumin fusion proteins of the invention to a protein, antigen or epitope. BIAcore kinetic analysis comprises analyzing the binding and dissociation of albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes or albumin fusion proteins, respectively, on their surface.
[0145] Antibodies that bind a Therapeutic protein corresponding to the Therapeutic protein portion of an albumin fusion protein may also be described or specified in terms of their binding affinity for a given protein or antigen, preferably the antigen which they specifically bind. Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-2 M, 10-2 M, 5 X 10-3 M, 10-3 M, 5 X 10-4 M, 10-4 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-5 M, 10-5 M, 5 10-6 A4, 10-6M, 5 X 10-7 M, 107 M, 5 X 10-8 M or 10-8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X
10r9M, 1 0.9 M, 5 X
1(110 M, 10110 M, 5 X 10-11 M, le M, 5 X 10-12 M, 1042 NI 5 X 10-13 M, 10-13 M, 5 X 1044 M, 10-14 M, 5 X 10115 M, or 10-15 M. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody. In addition, assays =

described herein (see Examples and Table 1) and otherwise known in the art may routinely be applied to measure the ability of albumin fusion proteins and fragments, variants and derivatives thereof to elicit biological activity and/or Therapeutic activity (either in vitro or in vivo) related to either the Therapeutic protein portion and/or albumin portion of the albumin fusion protein. Other methods will be known to the skilled artisan and are within the scope of the invention.
Albumin [0146] As described above, an albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion.
[0147] An additional embodiment comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are linked to one another by chemical conjugation.
[0148] The terms, human serum albumin (HSA) and human albumin (HA) are used interchangeably herein. The terms, "albumin and "serum albumin" are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
[0149] As used herein, "albumin" refers collectively to albumin protein or amino acid sequence, or an albumin fragment or variant, having one or moA functional activities (e.g., biological activities) of albumin. In particular, "albumin" refers to human albumin or fragments thereof (see for example, EP 201 239, EP 322 094 WO 97/24445, W095/23857) especially the mature form of human albumin as shown in Figure 1 and SEQ ID
NO: 1038, or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof.
[0150] In preferred embodiments, the human serum albumin protein used in the albumin fusion proteins of the invention contains one or both of the following sets of point mutations with reference to SEQ NO: 1038: Leu-407 to Ala, Leu-408 to Val, Val-409 to Ala, and Arg-410 to Ala; or Arg-410 to A, Lys-413 to Gin, and Lys-414 to Gln (see, e.g., International Publication No. W095/23857).
In even more preferred embodiments, albumin fusion proteins of the invention that contain one or both of above-described sets of point mutations have improved stability/resistance to yeast Yap3p proteolytic cleavage, allowing increased production of recombinant albumin fusion proteins expressed in yeast host cells.
[0151] As used herein, a portion of albumin sufficient to prolong the therapeutic activity or shelf-life of the Therapeutic protein refers to a portion of albumin sufficient in length or structure to stabilize or prolong the therapeutic activity of the protein so that the shelf life of the Therapeutic protein portion of the albumin fusion protein is prolonged or extended compared to the shelf-life in the non-fusion state. The albumin portion of the albumin fusion proteins may comprise the full length of the HA sequence as described above, or may include one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity. Such fragments may be of 10 or more amino acids in length or may include about 15, 20, 25, 30, 50, or more contiguous amino acids from the HA
sequence or may include part or all of specific domains of HA. For instance, one or more fragments of HA spanning the first two immunoglobulin-like domains may be used. In a preferred embodiment, the HA fragment is the mature form of HA.
[0152] The albumin portion of the albumin fusion proteins of the invention may be a variant of normal .HA. The Therapeutic protein portion of the albumin fusion proteins of the invention may also be variants of the Therapeutic proteins as described herein. The term "variants" includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
[0153] In particular, the albumin fusion proteins of the invention may include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419). The albumin may be derived from any vertebrate, especially any mammal, for example human, cow, sheep, or pig. Non-mammalian albumins include, but are not limited to, hen and salmon. The albumin portion of the albumin fusion protein may be from a different animal than the Therapeutic protein portion.
[0154] Generally speaking, an HA fragment or variant will be at least 100 amino acids long, preferably at least 150 amino acids long. The HA variant may consist of or alternatively comprise at least one whole domain of HA, for example domains 1 (amino acids 1-194 of SEQ ID NO: 1038), domain 2 (amino acids 195-387 of SEQ ID NO: 1038), domain 3 (amino acids 388-585 of SEQ ID NO: 1038), domains 1 and 2 (1-387 of SEQ ID
NO:

1038), domains 2 and 3 (195-585 of SEQ ID NO: 1038) or domains 1 and 3 (amino acids 1-194 of SEQ 1D NO: 1038 and amino acids 388-585 of SEQ ID NO: 1038). Each domain is itself made up of two homologous subdomains namely 1-105, 120-194, 195-291, 316-387, 388-491 and 512-585, with flexible inter-subdomain linker regions comprising residues Lys106 to Glu119, G1u292 to Va1315 and G1u492 to Ala511.
101551 Preferably, the albumin portion of an albumin fusion protein of the invention comprises at least one subdomain or domain of HA or conservative modifications thereof. If the fusion is based on subdomains, some or all of the adjacent linker is preferably used to link to the Therapeutic protein moiety.
Antibodies that Speccally bind Therapeutic proteins are also Therapeutic proteins 101561 The present invention also encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that specifically binds a Therapeutic protein disclosed in Table 1. It is specifically contemplated that the term "Therapeutic protein"
encompasses antibodies that bind a Therapeutic protein (e.g., as Described in column I of Table 1) and fragments and variants thereof. Thus an albumin fusion protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an antibody that binds a Therapeutic protein.
Antibody structure and background [0157] The basic antibody structural unit is known to comprise a tetramer. Each tetratner is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region f about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as kappa and lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, 1 gG, IgA, and IgE, respectively. See generally, Fundamental Immunology Chapters 3-5 (Paul, W., ed., 4th ed. Raven Press, N.Y.
(1998)). The variable regions of each light/heavy chain pair form the antibody binding site.
[0158] Thus, an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same.

[0159] The chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDR regions, in general, are the portions of the antibody which make contact with the antigen and determine its specificity. The CDRs from the heavy and the light chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chains variable regions comprise the domains FR!, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The variable regions are connected to the heavy or light chain constant region. The assignment of amino acids to each domain is in accordance with the defmitions of Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk J MoL Biol. 196:901-917 (1987); Chothia et al. Nature 342:878-883 (1989).
(0160] As used herein, "antibody" refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen (e.g., a molecule containing one or more CDR regions of an antibody). Antibodies that may correspond to a Therapeutic protein portion of an albumin fusion protein include, but are not limited to, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies (e.g., single chain Fvs), Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies specific to antibodies of the invention), and epitope-binding fragments of any of the above (e.g., VH
domains, VL
domains, or one or more CDR regions).
Antibodies that bind Therapeutic Proteins [0161] The present invention encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that binds a Therapeutic Protein (e.g., as disclosed in Table 1) or fragment or variant thereof.
(0162] Antibodies that bind a Therapeutic protein (or fragment or variant thereof) may be from any animal origin, including birds and mammals. Preferably, the antibodies are human, murine (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken antibodies. Most preferably, the antibodies are human antibodies. As used herein, "human" antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries and xenomice or other organisms that have been genetically engineered to produce human antibodies.
[0163] The antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1 , IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule. In preferred embodiments, the antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are IgG1 . In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are IgG2. In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are IgG4.
[0164] Most preferably the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain. Antigen-binding antibody fragments, including single-chain antibodies, may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains.
[0165] The antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of a Therapeutic protein or may be specific for both a Therapeutic protein as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360;
WO
92/05793; Tuft, et al., J. Immunol. 147:60-69 (1991); U.S. Patent Nos.
4,474,893; 4,714,681;
4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol. 148:1547-1553 (1992).
[0166] Antibodies that bind a Therapeutic protein (or fragment or variant thereof) may be bispecific or bifunctional which means that the antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann Clin. Exp.
Immunol. 79: 315-321 (1990), Kostelny et al. J Immunol. 148:1547 1553 (1992).
In addition, bispecific antibodies may be formed as "diabodies" (Holliger et al.
"Diabodiest: small bivalent and bispecific antibody fragments" PNAS USA 90:6444-6448 (1993)) or "Janusins"
(Traunecker et al. "Bispecific single chain molecules (Janusins) target cytotoxic lymphocytes on HIV infected cells" EMBO J 10:3655-3659 (1991) and Traunecker et al.
"Janusin: new molecular design for bispecific reagents" Int J Cancer Suppl 7:51-52 (1992)).
[0167] The present invention also provides albumin fusion proteins that comprise, fragments or variants (including derivatives) of an antibody described herein or known elsewhere in the art. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding a molecule of the invention, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions. Preferably, the variants (including derivatives) encode less than 50 amino acid substitutions, less than 40 amino acid substitutions, less than 30 amino acid substitutions, less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the reference VII domain, VHCDR1, VHCDR2, VHCDR3, VL domain, VLCDR1, VLCDR2, or VLCDR3. In specific embodiments, the variants encode substitutions of VHCDR3. In a preferred embodiment, the variants have conservative amino acid substitutions at one or more predicted non-essential =
amino acid residues.
[0168] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may be described or specified in terms of the epitope(s) or portion(s) of a Therapeutic protein which they recognize or specifically bind. Antibodies which specifically bind a Therapeutic protein or a specific epitope of a Therapeutic protein may also be excluded. Therefore, the present invention encompasses antibodies that specifically bind Therapeutic proteins, and allows for the exclusion of the same. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, binds the same epitopes as the unfused fragment or variant of that antibody itself.
[0169] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, or homolog of a Therapeutic protein are included. Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% sequence identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention. In specific embodiments, antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein cross-react with murine, rat and/or rabbit homologs of human proteins and the corresponding epitopes thereof. Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% sequence identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention. In a specific embodiment, the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and/or immunogenic polypeptides disclosed herein. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially identical cross reactivity characteristics compared to the fragment or variant of that particular antibody itself.

Further included in the present invention are antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide encoding a Therapeutic protein under stringent hybridization conditions (as described herein).
Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their binding affinity to a polypeptide of the invention. Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-2 M, 10-2 M, 5 X
10-3 M, 10-3 M, 5 X 104 M, 10-4 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-5 M, 10-5 M, 5 X 10-6 M, 10-6M, 5 X 10-7 M, 107 M, 5 X 10-8 M or 10-8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X 1019 M, i0 M, 5 X 104 M, 10-io M, 5 X 1041 M, 1041 M, 5 X 1042 M, 10-12 M, 5 X 1043 M, 10-13 M, 5 X 10-14 M, 10-14 M, 5 X 10-15 M, or 10-15 M. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody.
[0171] The invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of a Therapeutic protein as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein. In preferred embodiments, the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of a second antibody to an epitope of a Therapeutic protein. In other preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of a second antibody to an epitope of a Therapeutic protein by at least 95%, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
[0172] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may act as agonists or antagonists of the Therapeutic protein. For example, the present invention includes antibodies which disrupt the receptor/ligand interactions with the polypeptides of the invention either partially or fully. The invention features both receptor-specific antibodies and ligand-specific antibodies. The invention also features receptor-specific antibodies which do not prevent ligand binding but prevent receptor activation. Receptor activation (i.e., signaling) may be determined by techniques described herein or otherwise known in the art.
For example, receptor activation can be determined by detecting the phosphorylation (e.g., tyrosine or serine/threonine) of the receptor or its substrate by irrununoprecipitation followed by western blot analysis (for example, as described supra). In specific embodiments, antibodies are provided that inhibit ligand activity or receptor activity by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50% of the activity in absence of the antibody. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially similar characteristics with regard to preventing ligand binding and/or preventing receptor activation compared to an un-fused fragment or variant of the antibody that binds the Therapeutic protein.

101731 The invention also features receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand. Likewise, included in the invention are neutralizing antibodies which bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies which bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor. Further included in the invention are antibodies which activate the receptor. These antibodies may act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-mediated receptor activation, for example, by inducing dimerization of the receptor. The antibodies may be specified as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the Therapeutic proteins (e.g. as disclosed in Table 1). The above antibody agonists can be made using methods known in the art. See, e.g., PCT publication WO 96/40281; U.S.
Patent No.
5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res.

58(16):3668-3678 (1998); Harrop et al., J. Inummol. 161(4):1786-1794 (1998);
Thu et al., Cancer Res. 58(15):3209-3214 (1998); Yoon.et al., J. Inununol. 160(7):3170-3179 (1998);
Prat et al., J. Cell. Sci. 111(Pt2):237-247 (1998); Pitard et al., J. Immimol.
Methods 205(2):177-190 (1997); Liautard et al., Cytokine 9(4):233-241 (1997); Carlson et al., J. Biol.
Chem. 272(17):11295-11301 (1997); Taryman et al., Neuron 14(4):755-762 (1995);
Muller et al., Structure 6(9):1153-1167 (1998); Bartunek et al., Cytokine 8(1):14-20 (1996), In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, have similar or substantially identical agonist or antagonist properties as an un-fused fragment or variant of the antibody that binds the Therapeutic protein.
10174] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be used, for example, to purify, detect, and target Therapeutic proteins, including both in in vitro and in vivo diagnostic and therapeutic methods. For example, the antibodies have utility in immunoassays for qualitatively and quantitatively measuring levels of the Therapeutic protein in biological samples. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
Likewise, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, may be used, for example, to purify, detect, and target Therapeutic proteins, including both in vitro and in vivo diagnostic and therapeutic methods.
[0175] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody. For example, but not by way of limitation, the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, 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 may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc.
Additionally, the derivative may contain one or more non-classical amino acids. Albumin fusion proteins of the invention may also be modified as described above.
Methods of Producing Antibodies that bind Therapeutic Proteins [0176] The antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be generated by any suitable method known in the art. Polyclonal antibodies to an antigen-of-interest can be produced by various procedures well known in the art. For example, a Therapeutic protein may be administered to various host animals including, but not limited to, rabbits, mice, rats, etc. to induce the production of sera containing polyclonal antibodies specific for the antigen.
Various adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are also well known in the art.
101771 Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas (Elsevier, N.Y., 1981). The term "monoclonal antibody" as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
101781 Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. In a non-limiting example, mice can be immunized with a Therapeutic protein or fragment or variant thereof, an albumin fusion protein, or a cell expressing such a Therapeutic protein or fragment or variant thereof or albumin fusion protein. Once an immune response is detected, e.g., antibodies specific for the antigen are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC.
Hybridomns are selected and cloned by limited dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention. Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
101791 Accordingly, the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody. wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention.
[0180] Another well known method for producing both polyclonal and monoclonal human B cell lines is transformation using Epstein Barr Virus (EBV). Protocols for generating EBV-transformed B cell lines are commonly known in the art, such as, for example, the protocol outlined in Chapter 7.22 of Current Protocols in Immunology, Coligan et al., Eds., 1994, John Wiley & Sons, NY.
The source of B cells for transformation is commonly human peripheral blood, but B cells for transformation may also be derived from other sources including, but not limited to, lymph nodes, tonsil, spleen, tumor tissue, and infected tissues. Tissues are generally made into single cell suspensions prior to EBV transformation. Additionally, steps may be taken to either physically remove or inactivate T cells (e.g., by treatment with cyclosporin A) in B

cell-containing samples, because T cells from individuals seropositive for anti-EBV
antibodies can suppress B cell immortalization by EBV.
[0181] In general, the sample containing human B cells is innoculated with EBV, and cultured for 3-4 weeks. A typical source of EBV is the culture supernatant of the B95-8 cell line (ATCC #VR-1492). Physical signs of EBV transformation can generally be seen towards the end of the 3-4 week culture period. By phase-contrast microscopy, transformed cells may appear large, clear, hairy and tend to aggregate in tight clusters of cells. Initially, EBV lines are generally polyclonal. However, over prolonged periods of cell cultures, EBV
lines may become monoclonal or polyclonal as a result of the selective outgrowth of particular B cell clones. Alternatively, polyclonal EBV transformed lines may be subcloned (e.g., by limiting dilution culture) or fused with a suitable fusion partner and plated at limiting dilution to obtain monoclonal B cell lines. Suitable fusion partners for EBV
transformed cell lines include mouse myeloma cell lines (e.g., SP2/0, X63-Ag8.653), heteromyeloma cell lines (human x mouse; e.g, SPAM-8, SBC-H20, and CB-F7), and human cell lines (e.g., GM 1500, SKO-007, RPMI 8226, and KR-4). Thus, the present invention also provides a method of generating polyclonal or monoclonal human antibodies against polypeptides of the invention , or fragments thereof, comprising EBV-transformation of human B cells.
[0182] Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
[0183] For example, antibodies that bind to a Therapeutic protein can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In a particular embodiment, such phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene DI or gene VIII protein. Examples of phage display methods that can be used to make antibodies that bind to a Therapeutic protein include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et al., Gene 187 9-18 (1997); Burton et at., Advances in Immunology 57:191-280 (1994); PCT
application No.
PCT/GB91/01134; PCT publications WO 90/02809; WO 91/10737; WO 92/01047; WO
92/18619; WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Patent Nos.
5,698,426;
5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698;
5,427,908;
5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969,108.
[01841 As described in the above references, after phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below. For example, techniques to recombinantly produce Fab, Fab' and F(ab1)2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et at., BioTechniques 12(6):864-869 (1992); and Sawai et al., AJRI 34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988).
[01851 Examples of techniques which can be used to produce single-chain Fvs and antibodies include those described in U.S. Patents 4,946,778 and 5,258,498;
Huston et at., Methods in Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993);
and Skerra et al., Science 240:1038-1040 (1988). For some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use chimeric, humanized, or human antibodies. A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human inuntmoglobulin constant region. Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986);
Gillies et al., (1989) J. Inununol. Methods 125:191-202; U.S. Patent Nos. 5,807,715;
4,816,567; and 4,816397. Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-' human species and a framework regions from a human immunoglobulin molecule. Often, framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S.
Patent No.
5,585,089; Rieclunann et al., Nature 332:323 (1988)).
Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT
publication WO
91/09967; U.S. Patent Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; PadIan, Molecular Immunology 28(4/5):489-498 (1991);
Studnicka et al., Protein Engineering 7(6);805-814 (1994); Roguska. et al., PNAS 91:969-973 (1994)), and chain shuffling (U.S. Patent No. 5,565,332).
[0186]
Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences.
See also, U.S. Patent Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO
98/16654, WO 96/34096, WO 96/33735, and WO 91/10741.
[0187]
Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For example, the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy .and light chain genes. The mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production.
The modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring which express human antibodies. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all Or a portion of a polypeptide of the invention.
Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg and Huszar, Int. Rev. Immunol.
13:65-93 (1995). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., PCT
publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877; U.S. Patent Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825;
5,661,016;
5,545,806; 5,814,318; 5,885,793; 5,916,771; 5,939,598; 6,075,181; and 6,114,598.
In addition, companies such as Abgenix, Inc. (Freemont, CA) and Genpharm (San Jose, CA) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
[01881 Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection." In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et al., ho/technology 12:899-903 (1988)).
Polynudeotides Encoding Antibodies 101891 The invention further provides polynucleotides comprising a nucleotide sequence encoding an antibody and fragments thereof. The invention also encompasses polynucleotides that hybridize under stringent or alternatively, under lower stringency hybridization conditions, e.g., as defined supra, to polynucleotides that encode an antibody, preferably, that specifically binds to a Therapeutic protein, and more preferably, an antibody that binds to a polypeptide having the amino acid sequence of a "Therapeutic protein:X" as disclosed in the "SEQ ID NO:Z" column of Table 2.
101901 The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. For example, if the nucleotide sequence of the antibody is known, a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
[0191] Alternatively, a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA
clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art (See Example 107).
[0192] Once the nucleotide sequence and corresponding amino acid sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology, John Wiley & Sons, NY, to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions.
[0193] In a specific embodiment, the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the complementarity determining regions (CDRs) by methods that are well know in the art, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability. Using routine recombinant DNA
techniques, one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra. The framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia et al., J. Mol. Biol. 278: 457-479 (1998) for a listing of human framework regions). Preferably, the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention. Preferably, as discussed supra, one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds. Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art.
[0194] In addition, techniques developed for the production of "chimeric antibodies"
(Morrison et al., Proc. Natl. Acad. Sci. 81:851-855 (1984); Neuberger et al., Nature 312:604-608 (1984); Takeda et al., Nature 314:452-454 (1985)) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. As described supra, a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglzbulin constant region, e.g., humanized antibodies.
[01951 Alternatively, techniques described for the production of single chain antibodies (U.S. Patent No. 4,946,778; Bird, Science 242:423- 42 (1988);
Huston et al., Proc.
Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et al., Nature 334:544-54 (1989)) can be adapted to produce single chain antibodies. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli may also be used (Skerra etal., Science 242:1038- 1041 (1988)).
Recombinant Expression of Antibodies [0196] Recombinant expression of an antibody, or fragment, derivative or analog thereof, (e.g., a heavy or light chain of an antibody or a single chain antibody), requires construction of an expression vector containing a polynucleotide that encodes the antibody.

Once a polynucleotide encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably containing the heavy or light chain variable domain), of the invention has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art.
Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals.
These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter. Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036;
and U.S. Patent No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
[0197] The expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody.
Thus, the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody, operably linked to a heterologous promoter. In preferred embodiments for the expression of double-chained antibodies, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.

variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences;
insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
101991 In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited, to the E. coli expression vector pLTR278 (Ruther et al., EMBO J.
2:1791 (1983)), in which the antibody coding sequence may be ligated individually. into the vector in frame with the lac Z coding region so that a fusion protein is produced; plN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke &
Schuster, J. Biol.
Chem. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
[0200] In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).

[02011 In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts. (e.g., see Logan & Shenk, Proc. Natl. Acad. Sci. USA 81:355-359 (1984)). Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., Methods in Enzymol.
153:51-544(1987)).
[02021 In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
Such mammalian host cells include but are not limited to CHO, VERY, BHX., Hela, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 and Hs578Bst.
[02031 For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule. Such engimered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
[0204] A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad.
Sci. USA
48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes:
dhfr, which confers resistance to methotrexate (Wigler et al, Natl. Acad. Sci. USA 77:357 (1980);
O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA 78:2072 (1981)); neo, which confers resistance to the aminog,lycoside G-418 Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993);
. Mulligan, Science 260:926-932 (1993); and Morgan. and Anderson, Ann. Rev.
Biochem.
62:191-217 (1993); May, 1993, TM TECH 11(5):155-215 (1993)); and hygro, which confers resistance to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example, in Ausubel et at. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993);
Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990); and in Chapters 12 and 13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John Wiley & Sons, NY (1994); Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981).
[0205] The expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA
cloning, Vol.3.
(Academic Press, New York, 1987)). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., Mol.
Cell. Biol. 3:257 (1983)).
102061 Vectors which use glutamine synthase (GS) or DIER as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g. Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications:
W087/04462;
W086/05807; W089/01036; W089/10404; and W091/06657.
Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suppliers, including, for example Lonza Biologics, Inc. (Portsmouth, NH). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol.
Prog. 11:1 (1995).
[02071 The host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
Alternatively, a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986);
Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)). The coding sequences for the heavy and fight chains may comprise cDNA or genomic DNA.
102081 Once an antibody molecule of the invention has been produced by an animal, chemically synthesized, or recombinantly expressed, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. In addition, the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification.
Modifications of Antibodies [0209]
Antibodies that bind a Therapeutic protein or fragments or variants can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available. As described in Gentz et al., Proc.
Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the hemagglutinin tag (also called the "HA
tag"), which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the "flag" tag. =
[0210] The present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent. The antibodies can be used diagnostically to, for example, monitor the development or progression of a tumor as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen.
Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions. The detectable substance may be coupled or conjugated either directly to the antibody (or fragment thereof) or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 1251, 1311, 111In or 99Tc. Other examples of detectable substances have been described elsewhere herein.

Further, an antibody of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi. A
cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
[0212] The conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is nOt to be construed as limited to classical . chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, B-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I
(See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi etal., Int. Immunol., 6:1567-1574 (1994)), VEGI
(See, International Publication No. WO 99/23105), a thrombotic agent or an anti- angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lympholcines, interleukin-1 ("IL-1"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors.
102131 Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
(02141 Techniques for conjugating such therapeutic moiety to antibodies are well known. See, for example, Anion et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al.
(eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp.
623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A
Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", hmnunol. Rev. 62:119-58 (1982).
[0215] Alternatively, an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980.
[0216] An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
Antibody-albumin fusion [0217] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, antibodies that bind a Therapeutic protein disclosed in the "Therapeutic Protein X"
column of Table 1, or a fragment or variant thereof.
[0218] In specific embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the Vii domain.

In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VH
CDRs. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR1. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR2. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR3.
[0219] In specific embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL domain.
In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VL
CDRs. In other embodiments, the fragment = or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic Protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR1. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR2. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR3.
[0220] In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two, three, four, five, or six VH and/or VL CDRs.
[0221] In preferred embodiments, the fragment or variant of an antibody that immunospecifically binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, an scFv comprising the VH domain of the Therapeutic antibody, linked to the VL domain of the therapeutic antibody by a peptide linker such as (Gly4Ser)3(SEQ ID NO:1092).
immunophenotyping [0222] The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be utilized for immunophenotyping of cell lines and biological samples. Therapeutic proteins of the present invention may be useful as cell-specific markers, or more specifically as cellular markers that are differentially expressed at various stages of differentiation and/or maturation of particular cell types.
Monoclonal antibodies (or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) directed against a specific epitope, or combination of epitopes, will allow for the screening of cellular populations expressing the marker.
Various techniques can be utilized using monoclonal antibodies (or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, "panning" with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S. Patent 5,985,660; and Morrison et al., Cell, 96:737-49 (1999)). .
[0223] These techniques allow for the screening of particular populations of cells, such as might be found with hematological malignancies (i.e. minimal residual disease (MRD) in acute leukemic patients) and "non-self" cells in transplantations to prevent Graft-versus-Host Disease (GVHD). Alternatively, these techniques allow for the screening of hematopoietic stem and progenitor cells capable of undergoing proliferation and/or differentiation, as might be found in human umbilical cord blood.
Characterizing Antibodies that bind a Therapeutic Protein and Albumin Fusion Proteins Comprising a Fragment or Variant of an Antibody that binds a Therapeutic Protein [0224] The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be characterized in a variety of ways. In particular, Albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the antibody that binds a Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art.
[0225] Assays for the ability of the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Patent No. 5,223,409), on spores (e.g., Patent Nos. 5,571,698;
5,403,484; and 5,223,409), on plasmids (e.g., Cull et al., Proc. Natl. Acad.
Sci. USA
89:1865-1869 (1992)) or on phage (e.g., Scott and Smith, Science 249:386-390 (1990);
Devlin, Science 249:404-406(1990); Cwirla et al., Proc. NatL Acad. Sci. USA
87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)), The antibodies of the invention or albumin.
fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art.
[0226] The albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art.
102271 Immunoassays which can be used to analyze (inamunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA
(enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, inununodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York).
Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
[0228] Inununoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or TritoriX-100, 1% sodium deoxycholate, TIA
0.1% SDS, 0.15 M NaC1, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding an antibody of the invention or albumin fusion protein of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C, adding protein A and/or protein G sepharosrbeads (or beads coated with an appropriate anti-idiotypic antibody or anti-albumin antibody in the case when an albumin fusion protein comprising at least a fragment or variant of a Therapeutic antibody) to the cell lysate, incubating for about an hour or more at 40 degrees C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the antibody or albumin fusion protein of the invention to inununoprecipitate a particular antigen can be assessed by, e.g., western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody or albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.
[0229] Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20%
SDS-PAGE
depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween1120), applying the antibody or albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 1251) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.

ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the antibody or albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the antibody or albumin fusion proteins specifically bound to the antigen coating the well. In ELISAs the antibody or albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody or albumin fusion protein, respectively) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, antibody or the albumin fusion protein may be coated to the well. In this case, the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase).
One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.
[0231] The binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an antibody- or albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding assays. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 1251) with the antibody or albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen.
The affinity of the antibody or albumin fusion protein of the invention for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis. Competition with a second protein that binds the same protein, antigen or epitope as the antibody or albumin fusion protein, can also be determined using radioimmunoassays. In this case, the protein, antigen or epitope is incubated with an antibody or albumin fusion protein of the invention conjugated to a labeled compound (e.g., 3H or )25I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
[0232] In a preferred embodiment, BIAcore kinetic analysis is used to determine the binding on and off rates of antibody or albumin fusion proteins of the invention to a protein, antigen or epitope. BlAcore kinetic analysis comprises analyzing the binding and dissociation of antibodies, albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes, antibodies or albumin fusion proteins, respectively, on their surface.
Therapeutic Uses 10233] The present invention is further directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more of the disclosed diseases, disorders, or conditions. Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein), nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein), albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, and nucleic acids encoding such albumin fusion proteins. The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein. The treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions. antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.

102341 In a specific and preferred embodiment, the present invention is directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions., and/or as described elsewhere herein. Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (e.g., antibodies directed to the full length protein expressed on the cell surface of a mammalian cell; antibodies directed to an epitope of a Therapeutic protein and nucleic acids encoding antibodies of the invention (including fragments, analogs = and derivatives thereof and anti-idiotypic antibodies as described herein). The antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein. The treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions. Antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
[0235] A summary of the ways in which the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be used therapeutically includes binding Therapeutic proteins locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below. Armed with the teachings provided herein, one of ordinary skill in the art will know how to use the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein for diagnostic, monitoring or therapeutic purposes without undue experimentation.

[02361 The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
[0237] The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents).

Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred.
Thus, in a preferred embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis.
[0238] It is preferred to use high affinity and/or potent in vivo inhibiting and/or neutralizing antibodies against Therapeutic proteins, fragments .or regions thereof, (or the albumin fusion protein correlate of such an antibody) for both immunoassays directed to and therapy of disorders related to polynucleotides or polypeptides, including fragments thereof, of the present invention. Such antibodies, fragments, or regions, will preferably have an affinity for polynucleotides or polypeptides of the invention, including fragments thereof.
Preferred binding affinities include dissociation constants or Kd's less than 5 X 10-2 M, 10-2 M, 5 X 10-3 M, 10-3 M, 5 X 104 M, 104 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-5 M, 10-5 M, 5 X 10-6 M, 10-6M, 5 X 10-7 M, 107 M, 5 X 10-8 M or 10-8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-9 M, 10-9 M, 5 X 10-1 M, 10-10 M, 5 X 10-"
10-11 M, 5 X 10-12 M, 10-12 M, 5 X 10-13 M, 10-13 M, 5 X 1044 M, 1044 M, 5 X
1045 M, or 10-15 m.
Gene Therapy [0239] In a specific embodiment, nucleic acids comprising sequences encoding antibodies that bind therapeutic proteins or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect.
[0240] Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described in more detail elsewhere in this application.
Demonstration of Therapeutic or Prophylactic Activity [0241] The compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans. For example, in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample. The effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays. In accordance with the invention, in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed.
Therapeutic/Prophylactic Administration and Composition [0242] The invention provides methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a compound or pharmaceutical composition of the invention. In a preferred embodiment, the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects). The subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human.
[0243]
Formulations and methods of administration that can be employed when the compound comprises a nucleic acid or an immunoglobulin are described above;
additional appropriate formulations and routes of administration can be selected from among those described herein below.
[02441 Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of introduction include but are not limited to intradennal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the pharmaceutical compounds or compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ornmaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
[02451 In a specific embodiment, it may be desirable to administer the pharmaceutical compounds or compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a protein, including an antibody, of the invention, care must be taken to use materials to which the protein does not absorb.
[0246] In another embodiment, the compound or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990);
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); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.) [0247] In yet another embodiment, the compound or composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra;

Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980);
Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J.Neurosurg. 71:105 (1989)). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
[0248] Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
[0249] In a specific embodiment where the compound of the invention is a nucleic acid encoding a protein, the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Patent No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox- like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. ,Natl. Acad. Sci.
USA 88:1864-1868 (1991)), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
[0250] The present invention also provides pharmaceutical compositions.
Such compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier. In a specific embodiment, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile 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. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients 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. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
102511 In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
[0252] The compounds of the invention can be formulated as neutral or salt forms.
Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.

[0253] The amount of the compound of the invention which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
[0254] For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight. Preferably, the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight. Generally, human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible. Further, the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.
Diagnosis and Imaging [0255] Labeled antibodies and derivatives and analogs thereof that bind a Therapeutic protein (or fragment or variant thereof) (including albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein), can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders, and/or conditions associated with the aberrant expression and/or activity of Therapeutic protein. The invention provides for the detection of aberrant expression of a Therapeutic protein, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein expression level compared to the standard expression level is indicative of aberrant expression.
[0256] The invention provides a diagnostic assay for diagnosing a disorder, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the Therapeutic protein or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein, and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein gene expression level compared to the standard expression level is indicative of a particular disorder. With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
(0257]
Antibodies of the invention or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein can be used to assay protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen etal., J. Cell. Biol. 101:976-985 (1985); Jalkanen et al., J. Cell . Biol. 105:3087-3096 (1987)). Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (311), indium (1121n), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0258] One facet of the invention is the detection and diagnosis of a disease or disorder associated with aberrant expression of a Therapeutic protein in an animal, preferably a mammal and most preferably a human. In one embodiment, diagnosis comprises:
a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled molecule which specifically binds to the polypeptide of interest; b) waiting for a time interval following the administering for permitting the labeled molecule to preferentially concentrate at sites in the subject where the Therapeutic protein is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled molecule in the subject, such that detection of labeled molecule above the background level indicates that the subject has a particular disease or disorder associated with aberrant expression of the therapeutic protein.

Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.
[0259] It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. The labeled antibody, antibody fragment, or albumin fusion protein comprising at least a fragment or variant of an antibody that binds a Therapeutic protein will then preferentially accumulate at the location of cells which contain the specific Therapeutic protein. In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments." (Chapter 13 in Tumor Imaging:
The Radiochemical Detection of Cancer, S.W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
[0260] Depending on several variables, including the type of label used and the mode of administration, the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours.
In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days.
[0261] In an embodiment, monitoring of the disease or disorder is carried out by repeating the method for diagnosing the disease or disease, for example, one month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc.
[0262] Presence of the labeled molecule can be detected in the patient using methods known in the art for in vivo scanning. These methods depend upon the type of label used.
Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
[0263] In a specific embodiment, the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S.
Patent No. 5,441,050). In another embodiment, the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument.
In another embodiment, the molecule is labeled with a positron emitting metal and is detected in the patent using positron emission-tomography. In yet another embodiment, the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI). Antibodies that specifically detect the albumin fusion protein but not albumin or the therapeutic protein alone are a preferred embodiment. These can be used to detect the albumin fusion protein as described throughout the specification.
Kits [0264] The present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises an antibody, preferably a purified antibody, in one or more containers. In a specific embodiment, the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit. Preferably, the kits of the present invention further comprise a control antibody which does not react with the polypeptide of interest. In another specific embodiment, the kits of the present invention contain a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody- may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate).
[0265] In another specific embodiment of the present invention, the kit is a diagnostic kit for use in 'screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides. Such a kit mayqnclude a control antibody that does not react with the polypeptide of interest. Such a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody. Further, such a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry). In specific embodiments, the kit may include a recombinantly produced or chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support.
[0266] In a more specific embodiment the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached. Such a kit may also include a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
[0267] In an additional embodiment, the invention includes a diagnostic kit for use in screening serum containing antigens of the polypeptide of the invention. The diagnostic kit includes a substantially isolated antibody specifically immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody. In one embodiment, the antibody is attached to a solid support. In a specific embodiment, the antibody may be a monoclonal antibody.
The detecting means of the kit may include a second, labeled monoclonal antibody.
Alternatively, or in addition, the detecting means may include a labeled, competing antigen.
[0268] In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention.
After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support. The reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined. Typically, the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, MO).
[0269] The solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plate or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s).
[0270] Thus, the invention provides an assay system or kit for carrying out this diagnostic method. The kit generally includes a support with surface-bound recombinant antigens, and a reporter-labeled anti-human antibody for detecting surface-bound anti-antigen antibody.
Albumin Fusion Proteins 102711 The present invention relates generally to albumin fusion proteins and methods of treating, preventing, or ameliorating diseases or disorders. As used herein, "albumin fusion protein" refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof). An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or to one another. The Therapeutic protein and albumin protein, once part of the albumin fusion protein, may each be referred to as a "portion", "region" or "moiety" of the albumin fusion protein.
[0272] In a preferred embodiment, the invention provides an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 1 or Table 2.
Polynucleotides encoding these albumin fusion proteins are also encompassed by the invention.
102731 Preferred albumin fusion proteins of the invention, include, but are not limited to, albumin fusion proteins encoded by a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof); a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof) generated as described in Table 1, Table 2 or in the Examples; or a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof), further comprising, for example, one or more of the following elements: (1) a functional self-replicating vector (including but not limited to, a shuttle vector, an expression vector, an integration vector, and/or a replication system), (2) a region for initiation of transcription (e.g., a promoter region, such as for example, a regulatable or inducible promoter, a constitutive promoter), (3) a region for termination of transcription, (4) a leader sequence, and (5) a selectable marker.
[0274] In one embodiment, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein (e.g., as described in Table 1) and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein. In preferred embodiments, the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
[0275] In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin. In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin. In preferred embodiments, the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein.
[0276] In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin. In preferred embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin.
[0277]
Preferably, the albumin fusion protein comprises HA as the N-terminal portion, and a Therapeutic protein as the C-terminal portion. Alternatively, an albumin fusion protein comprising HA as the C-terminal portion, and a Therapeutic protein as the N-terminal portion may also be used.
[0278] In other embodiments, the albumin fusion protein has a Therapeutic protein fused to both the N-terminus and the C-terminus of albumin. In a preferred embodiment, the Therapeutic proteins fused at the N- and C- termini are the same Therapeutic proteins. In an alternative preferred embodiment, the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins. In another preferred embodiment, the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins which may be used to treat or prevent the same or a related disease, disorder, or condition (e.g. as listed in the "Preferred Indication Y" column of Table 1). In another preferred embodiment, the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins which may be used to treat, ameliorate, or prevent diseases or disorders (e.g. as listed in the "Preferred Indication Y"
column of Table 1) which are known in the art to commonly occur in patients simultaneously, concurrently, or consecutively, or which commonly occur in patients in association with one another.
[0279] Exemplary fusion proteins of the invention containing multiple Therapeutic protein portions fused at the N- and C- termini of albumin include, but are not limited to, GCSF-HSA-EPO, EPO-HSA-GCSF, IFNalpha-HSA-1L2, 1L2-HSA-IFNalpha, GCSF-HSA-112, 1L2-HSA-GCSF, 1L2-HSA-EPO, EPO-HSA-1L2, 1L3-HSA-EPO, EPO-HSA-1L3, GCSF-HSA-GMCSF, GMCSF-HSA-GCSF, 1L2-HSA-GMCSF, GMCSF-HSA-1L2, PTH-HSA-Calcitonin, Calcitonin-HSA-PTH, PTH-PTH-HSA-Calcitonin, Calcitonin-HSA-PTH-PTH, PTH-Calcitonin-HSA-PTH, or PTH-HSA-Calcitonin-PTH.
[0280] Albumin fusion proteins of the invention encompass proteins containing one, two, three, four, or more molecules of a given Therapeutic protein X or variant thereof fused to the N- or C- terminus of an albumin fusion protein of the invention, and/or to the N- and/or C- terminus of albumin or variant thereof. Molecules of a given Therapeutic protein X or variants thereof may be in any number of orientations, including, but not limited to, a 'head to head' orientation (e.g., wherein the N-terminus of one molecule of a Therapeutic protein X is fused to the N-terminus of another molecule of the Therapeutic protein X), or a 'head to tail' orientation (e.g., wherein the C-terminus of one molecule of a Therapeutic protein X is fused to the N-terminus of another molecule of Therapeutic protein X).
[0281] In one embodiment, one, two, three, or more tandemly oriented Therapeutic protein X polypeptides (or fragments or variants thereof) are fused to the N-or C- terminus of an albumin fusion protein of the invention, and/or to the N- and/or C-terminus of albumin or variant thereof.
[0282] In a specific embodiment, one, two, three, four, five, or more tandemly oriented molecules of PTH are fused to the N- or C-terminus of albumin or variant thereof.
For example, one, two, three, four, five, or more tandemly oriented molecules of PTH
(including, but not limited to, molecules of PTH comprising, or alternatively consisting of, amino acids 1 to 34) are fused to the N- or C-terminus of albumin or variant thereof.
Exemplary fusion proteins of the invention containing multiple protein portions of PTH, include, but are not limited to, PTH-PTH-HSA, HSA-PTH-PTH, PTH-PTH-PTH-HSA, HSA-PTH-PTH-PTH, PTH-PTH-PTH-PTH-HSA, or HSA-PTH-PTH-PTH-PTH.

[02831 In another specific embodiment, one, two, three, four, five, or more tandemly oriented molecules of GLP-1 are fused to the N- or C-terminus of albumin or variant thereof'.
For example, one, two, three, four, five, or more tandemly oriented molecules of GLP-1 (including, but not limited to, molecules of GLP-1 comprising, or alternatively consisting of, amino acids 7 to 36, with residue 8 being mutated from an Alanine to a Glycine) (See for Example, the mutants disclosed in U.S. Patent No. 5,545,618, are fused to the N- or C-terminus of albumin or variant thereof.
Exemplary fusion proteins of the invention containing multiple protein portions of GLP-1, include, but are not limited to, GL1-GLP1-HSA, HSA-GLP1-GLP1, GLP1mutant-GLP1mutant-HSA, HSA-GLP1mutant-GLP1mutant, GLP1mutant-GLP1-HSA, HSA-GLP1mutant-GLP 1, GLP1-GLP1mutant-HSA, or HSA-GLP1-GLP1mutant. Particularly preferred embodiments are GLP-1 tandem fusions such as construct ID #3070 and the protein encoded by such constuct.
[0284] Albumin fusion proteins of the invention further encompass proteins containing one, two, three, four, or more molecules of a given Therapeutic protein X or variant thereof fused to the N- or C- terminus of an albumin fusion protein of the invention, and/or to the N- and/or C- terminus of albumin or variant thereof, wherein the molecules are joined through peptide linkers. Examples include those peptide linkers described in U.S. Pat.
No. 5,073,627.
Albumin fusion proteins comprising multiple Therapeutic protein X polypeptides separated by peptide linkers may be produced using conventional recombinant DNA technology. Linkers are particularly important when fusing a small peptide to the large HSA molecule. The peptide itself can be a linker by fusing tandem copies of the peptide (see for example GLP-1) or other known linkers can be used.
Constructs that incorporate linkers are described in Table 2 or are apparent when examining SEQ ID NO:Y.
[02851 Further, albumin fusion proteins of the invention may also be produced by fusing a Therapeutic protein X or variants thereof to the N-terminal and/or C-tenninal of albumin or variants thereof in such a way as to allow the formation of intramolecular and/or intermolecular multimeric forms. In one embodiment of the invention, albumin fusion proteins may be in monomeric or multimeric forms (i.e., dimers, trimers, tetramers and higher multimers). In a further embodiment of the invention, the Therapeutic protein portion of an albumin fusion protein may be in Monomeric form or multimeric form (i.e., dimers, trimers, tetramers and higher multimerg). In a specific embodiment, the Therapeutic protein portion of an albumin fusion protein is in multimeric form (i.e., dimers, trimers, tetramers and higher multimers), and the albumin protein portion is in monomeric form.
[0286] In addition to albumin fusion protein in which the albumin portion is fused N-terminal and/or C-terminal of the Therapeutic protein portion, albumin fusion proteins of the invention may also be produced by inserting the Therapeutic protein or peptide of interest (e.g., a Therapeutic protein X as disclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof) into an internal region of HA. For instance, within the protein sequence of the HA molecule a number of loops or turns exist between the end and beginning of a-helices, which are stabilized by disulphide bonds. The loops, as determined from the crystal structure of HA (PDB identifiers IA06, 1BJ5, 1BKE, 1BMO, 1E7E to 1E71 and lUOR) for the most part extend away from the body of the molecule.
These loops are useful for the insertion, or internal fusion, of therapeutically active peptides, particularly those requiring a secondary structure to be functional, or Therapeutic proteins, to essentially generate an albumin molecule with specific biological activity.
[0287] Loops in human albumin structure into which peptides or polypeptides may be inserted to. generate albumin fusion proteins of the invention include: Va154-Asn61, Thr76-Asp89, Ala92-G1u100, G1n170-Ala176, His 247 - G1u252, Glu 266 - G1u277, Glu His288, A1a362-G1u368, Lys439-Pro447, Va1462-Lys475, Thr478-Pro486, and Lys560-Thr566. In more preferred embodiments, peptides or polypeptides are inserted into the Va154-Asn61, Gln170-A1a176, and/or Lys560-Thr566 loops of mature human albumin (SEQ
ID NO:1038).
[0288] Peptides to be inserted may be derived from either phage display or synthetic peptide libraries screened for specific biological activity or from the active portions of a molecule with the desired function. Additionally, random peptide libraries may be generated within particular loops or by insertions of randomized peptides into particular loops of the HA molecule and in which all possible combinations of amino acids are represented.
[0289] Such library(s) could be generated on HA or domain fragments of HA
by one of the following methods:
[0290] randomized mutation of amino acids within one or more peptide loops of HA
or HA domain fragments. Either one, more or all the residues within a loop could be mutated in this manner;
[0291] replacement of, or insertion into one or more loops of HA or HA
domain fragments (i.e., internal fusion) of a randomized peptide(s) of length Xõ
(where X is an amino acid and n is the number of residues;
[0292] N-, C- or N- and C- terminal peptide/protein fusions in addition to (a) and/or (b).
[0293] The HA or HA domain fragment may also be made multifunctional by gaffing the peptides derived from different screens of different loops against different targets into the same HA or HA domain fragment.
[0294] In preferred embodiments, peptides inserted into a loop of human serum albumin are peptide fragments or peptide variants of the Therapeutic proteins disclosed in Table 1. More particularly, the invention encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids in length inserted into a loop of human serum albumin.
The invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the N-terminus of human serum albumin. The invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the C-terminus of human serum albumin. For example, short peptides described in Table 1 and 2 (e.g., Therapeutic Y) can be inserted into the albumin loops.
[0295] Generally, the albumin fusion proteins of the invention may have one HA-derived region and one Therapeutic protein-derived region. Multiple regions of each protein, however, may be used to make an albumin fusion protein of the invention. Similarly, more than one Therapeutic protein may be used to make an albumin fusion protein of the invention. For instance, a Therapeutic protein may be fused to both the N- and C-terminal ends of the HA. In such a configuration, the Therapeutic protein portions may be the same or different Therapeutic protein molecules. The structure of bifunctional albumin fusion proteins may be represented as: X-HA-Y or Y-HA-X.
[0296] For example, an anti-BLySTM scFv-HA-IFNa-2b fusion may be prepared to modulate the immune response to IFNa-2b by anti-BLySTm scFv. An alternative is making a bi (or even multi) functional dose of HA-fusions e.g. HA-1FNa-2b fusion mixed with HA-anti-BLySTm scFv fusion or other HA-fusions in various ratio's depending on function, half-life etc.
[0297] Bi- or multi-functional albumin fusion proteins may also be prepared to target the Therapeutic protein portion of a fusion to a target organ or cell type via protein or peptide at the opposite terminus of HA.
[0298] As an alternative to the fusion of known therapeutic molecules, the peptides could be obtained by screening libraries constructed as fusions to the N-, C-or N- and C-termini of HA, or domain fragment of HA, of typically 6, 8, 12, 20 or 25 or Xõ
(where X is an amino acid (aa) and n equals the number of residues) randomized amino acids, and in which all possible combinations of amino acids were represented. A particular advantage of this approach is that the peptides may be selected in situ on the HA molecule and the properties of the peptide would therefore be as selected for rather than, potentially, modified as might be the case for a peptide derived by any other method then being attached to HA.
[0299] Additionally, the albumin fusion proteins of the invention may include a linker peptide between the fused portions to provide greater physical separation between the moieties and thus maximize the accessibility of the Therapeutic protein portion, for instance, . for binding to its cognate receptor. The linker peptide may consist of amino acids such that it =
is flexible or more rigid.
[0300] The linker sequence may be cleavable by a protease or chemically to yield the growth hormone related moiety. Preferably, the protease is one which is produced naturally by the host, for example the S. cerevisiae protease kex2 or equivalent proteases.
[0301] Therefore, as described above, the albumin fusion proteins of the invention may have the following formula R1 -L-R2; R2-L-R1; or R1-L-R2-L-R1, wherein R1 is at least one Therapeutic protein, peptide or polypeptide sequence, and not necessarily the same Therapeutic protein, L is a linker and R2 is a serum albumin sequence.
[0302] In preferred embodiments, Albumin fusion proteins of the invention comprising a Therapeutic protein have extended shelf life compared to the shelf life the same Therapeutic protein when not fused to albumin. Shelf-life typically refers to the time period over which the therapeutic activity of a Therapeutic protein in solution or in some other storage formulation, is stable without undue loss of therapeutic activity.
Many of the Therapeutic proteins are highly labile in their unfused state. As described below, the typical shelf-life of these Therapeutic proteins is markedly prolonged upon incorporation into the albumin fusion protein of the invention.
[0303] Albumin fusion proteins of the invention with "prolonged" or "extended"

shelf-life exhibit greater therapeutic activity relative to a standard that has been subjected to the same storage and handling conditions. The standard may be the unfused full-length Therapeutic protein. When the Therapeutic protein portion of the albumin fusion protein is an analog, a variant, or is otherwise altered or does not include the complete sequence for that protein, the prolongation of therapeutic activity may alternatively be compared to the unfused equivalent of that analog, variant, altered peptide or incomplete sequence. As an example, an albumin fusion protein of the invention may retain greater than about 100% of the therapeutic activity, or greater than about 105%, 110%, 120%, 130%, 150% or 200% of the therapeutic activity of a standard when subjected to the same storage and handling conditions as the standard when compared at a given time point.
[0304] Shelf-life may also be assessed in terms of therapeutic activity remaining after storage, normalized to therapeutic activity when storage began. Albumin fusion proteins of the invention with prolonged or extended shelf-life as exhibited by prolonged or extended therapeutic activity may retain greater than about 50% of the therapeutic activity, about 60%, 70%, 80%, or 90% or more of the therapeutic activity of the equivalent unfused Therapeutic protein when subjected to the same conditions. For example, as discussed in Example 38, an albumin fusion protein of the invention comprising hGH fused to the full length HA sequence may retain about 80% or more of its original activity in solution for periods of up to 5 weeks or more under various temperature conditions.
Expression of Fusion Proteins [0305] The albumin fusion proteins of the invention may be produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line. Preferably, the polypeptide is secreted from the host cells.
[0306] A
particular embodiment of the invention comprises a DNA construct encoding a signal sequence effective for directing secretion in yeast, particularly a yeast-derived signal sequence (especially one which is homologous to the yeast host), and the fused molecule of the first aspect of the invention, there being no yeast-derived pro sequence between the signal and the mature polypeptide.
[0307] The Saccharomyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence.
[0308]
Conjugates of the kind prepared by Poznansky et al., (FEBS Lett. 239:18 (1988)), in which separately-prepared polypeptides are joined by chemical cross-linking, are =
not contemplated.
[0309] The present invention also includes a cell, preferably a yeast cell transformed to express an albumin fusion protein of the invention. In addition to the transformed host cells themselves, the present invention also contemplates a culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away. Many expression systems are known and may be used, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
[03101 Preferred yeast strains to be used in the production of albumin fusion proteins are I)88, DXY1 and BX210. D88 [leu2-3, leu2-122, canl, pral, ubc4] is a derivative of parent strain AH22his+ (also known as DB1; see, e.g., Sleep et at.
Biotechnology 8:42-46 (1990)). The strain contains a leu2 mutation which allows for auxotropic selection of 2 micron-based plasmids that contain the LEU2 gene. D88 also exhibits a derepression of PRB1 in glucose excess. The PRB1 promoter is normally controlled by two checkpoints that monitor glucose levels and growth stage. The promoter is activated in wild type yeast upon glucose depletion and entry into stationary phase. Strain 1)88 exhibits the repression by glucose but maintains the induction upon entry into stationary phase. The PRA1 gene encodes a yeast vacuolar protease, YscA endoprotease A, that is localized in the ER. The UBC4 gene is in the ubiquitination pathway and is involved in targeting short lived and abnormal proteins for ubiquitin dependant degradation. Isolation of this ubc4 mutation was found to increase the copy number of an expression plasmid in the cell and cause an increased level of expression of a desired protein expressed from the plasmid (see, e.g., International Publication No. W099/00504)-(03111 DXY1, a derivative of 1)88, has the following genotype: [leu2-3, leu2-122, can!, pral , ubc4, ura3::yap3]. In addition to the mutations isolated in D88, this strain also has a knockout of the YAP3 protease. This protease causes cleavage of mostly di-basic residues (RR., RK, KR, KR) but can also promote cleavage at single basic residues in proteins. Isolation of this yap3 mutation resulted in higher levels of full length HSA
production (see, e.g., U.S. Patent No. 5,965,386 and Kerry-Williams et al., Yeast 14:161-169 (1998)).

=
[03121 BXP10 has the following genotype: leu2-3, leu2-122, can!, pra 1 , ubc4, ura3, yap3::URA3, lys2, hsp150::LYS2, pmt1::URA3. In addition to the mutations isolated in DXY1, this strain also has a knockout of the PMT1 gene and the HSP150 gene.
The PMT1 gene is a member of the evolutionarily conserved family of dolichyl-phosphate-D-marmose protein 0-mannosylfransferases (Pmts). The transmembrane topology of Pmtlp suggests that it is an integral membrane protein of the endoplasmic reticulum with a role in 0-linked glycosylation. This mutation serves to reduce/eliminate 0-linked glycosylation of HSA
fusions (see, e.g., International Publication No. W000/44772).
Studies revealed that the Hsp150 protein is inefficiently separated from rHA by ion exchange chromatography. The mutation in the HSP150 gene removes a potential contaminant that has proven difficult to remove by standard purification techniques. See, e.g., U.S. Patent No. 5,783,423.
[03131 The desired protein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid. The yeasts are transformed with a coding sequence for the desired protein in. any of the usual ways, for example electroporation. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) Methods Enzymol. 194, 182.
[03141 Successfully transformed cells, i.e., cells that contain a DNA
construct of the present invention, can be identified by well known techniques. For example, cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide. Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975)J.
Mol. Biol.
98, 503 or Berent et al. (1985) Biotech. 3, 208. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
[03151 Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, 7RP1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
[0316] Preferred vectors for making albumin fusion proteins for expression in yeast include pPPC0005, pScCHSA, pScNHSA, and pC4:HSA which are described in. detail in =
Example 1. Figure 2 shows a map of the pPPC0005 plasmid that can be used as the base vector into which polynucleotides encoding Therapeutic proteins may be cloned to form HA-fusions. It contains a PRBI S. cerevisiae promoter (PRB1p), a Fusion leader sequence (FL), DNA encoding HA (rHA) and an ADH1 S. cerevisiae terminator sequence. The sequence of the fusion leader sequence consists of the first 19 amino acids of the signal peptide of human serum albumin (SEQ ID NO:1094) and the last five amino acids of the mating factor alpha 1 promoter (SLDKR, see EP-A-387 319).
[0317] The plasmids, pPPC0005, pScCHSA, pScNHSA, and pC4:HSA were deposited on April 11, 2001 at the American Type Culture Collection, 10801 University Boulevard, Manassas, Virginia 20110-2209 and given accession numbers ATCC PTA-3278, PTA-3276, PTA-3279, and PTA-3277, respectively. Another vector useful for expressing an albumin fusion . protein in yeast the pSAC35 vector which is described in Sleep et al., BioTeclmology 8:42 (1990), [03181 Another yeast promoter that can be used to express the albumin fusion protein is the MET25 promoter. See, for example, Dominik Mumburg, Rolf Muller and Martin Funk. Nucleic Acids Research, 1994, Vol. 22, No. 25, pp. 5767-5768. The Met25 promoter is 383 bases long (bases ¨382 to ¨1) and the genes expressed by this promoter are also known as Met15, Met17, and YLR303W. A preferred embodiment uses the sequence below, where, at the 5' end of the sequence below, the Not 1 site used in the cloning is underlined and at the 3' end, the ATG start codon is underlined:
GeGGCCGCCGGATGCAAGGGTTCGAATCCCTTAGCTCTCATTATITITTGCTTTIT
CTCTTGAGGTCACATGATCGCAAAATGGCAAATGGCACGTGAAGCTGTCGATATT
GGGGAACTGTGGTGGITGGCAAATGACTAATTAAGITAGTCAAGGCGCCATCCTC
ATGAAAACTGTGTAACATAATAACCGAAGTGTCGAAAAGGTGGCACCTTGTCCA
ATTGAACACGCTCGATGAAAAAAATAAGATATATATAAGGTTAAGTAAAGCGTC
TGTTAGAAAGGAAGTMTCCMTTCTTGCTCTCTTGTCITITCAT;CTACTATTTC
CTTCGTGTAATACAGGGTCGTCAGATACATAGATACAATTCTATT'ACCCCCATCC
ATACAATG (SEQ ID NO: 2138) [03191 A variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA
segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules.

[0320] Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. The DNA segment, generated by endonuclease restriction digestion, is treated with bacteriophage T4 DNA
polymerase or E.
coli DNA polymerase I, enzymes that remove protruding, gamma-single-stranded termini with their 3' 5'-exonucleolytic activities, and fill in recessed 3'-ends with their polymerizing activities.
[0321] The combination of these activities therefore generates blunt-ended DNA
segments. The blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase. Thus, the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA
segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA
segment.
[0322] Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, CT, USA.
[0323] A desirable way to modify the DNA in accordance with the invention, if, for example, HA variants are to be prepared, is to use the polymerase chain reaction as disclosed by Said et a/. (1988) Science 239, 487-491. In this method the DNA to be enzymatically amplified is flanked by two specific oligonucleotide primers which themselves become incorporated into the amplified DNA. The specific primers may contain restriction endonuclease recognition sites which can be used for cloning into expression vectors using methods known in the art.
[0324] Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin fusion proteins are Pichia (Hansenula), Saccharomyces, Kluyveromyces, Candida, Torulopsis, Torulaspora, Schizosaccharomyces, Citeromyces, Pachysolen, Debaromyces, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
[0325] Examples of Kluyveromyces spp. are K fragilis, K lactis and K.
marxianus. A
suitable Torulaspora species is T. delbrueckii. Examples of Pichia (Hansenula) spp. are P.

dngusta (formerly H. polymorpha), P. anomala (formerly H. anomala) and P.
pastoris.
Methods for the transformation of S. cerevisiae are taught generally in EP 251 744, EP 258 067 and WO 90/01063.
[0326] Preferred exemplary species of Saccharomyces include S.
cerevisiae, S.
italicus, S. diastaticus, and Zygosaccharomyces rouxii. Preferred exemplary species of Kluyveromyces include K fragilis and K lactis. Preferred exemplary species of Hansenula include H. polymorpha (now Pichia angusta), H. anomala (now Pichia anomala), and Pichia capsulata. Additional preferred exemplary species of Pichia include P.
pastorif. Preferred exemplary species of Aspergillus include A. niger and A. nidulans. Preferred exemplary species of Yamowia include Y. lipolytica. Many preferred yeast species are available from the ATCC. For example, the following preferred yeast species are available from the ATCC
and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession No. 4022731);
Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp150 mutant (ATCC
Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmtl mutant (ATCC Accession No. 4023792); SaCcharomyces cerevisiae Hansen, teleomorph (ATCC Accession Nos. 20626; 44773; 44774; and 62995); Saccharomyces diastaticus Andrews et Gilliland ex van der Walt, teleomorph (ATCC Accession No. 62987);
Kluyveromyces lactis (Dombrowski) van der Walt, teleomorph (ATCC Accession No.

76492); Pichia angusta (Teunisson et al.) Kurtzman, teleomorph deposited as Hansenula polymorpha de Morals et Maia, teleomorph (ATCC Accession No. 26012);
Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 9029); Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 16404); Aspergillus nidulans (Eidam) Winter, anamorph (ATCC Accession No. 48756); and Yarrowia lipolytica (Wickerham et al.) van der Walt et von Aix, teleomorph (ATCC Accession No. 201847).
[0327] Suitable promoters for S. cerevisiae include those associated with the PGKI
gene, GAL1 or GAL10 genes, CYCI, PH05, TRPI, ADHI, ADH2, the genes for glyceraldehyde-3-phosphate dehycirogenase, hexolcinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoglucose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5' regulatory regions with parts of 5' regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).

[0328] Convenient regulatable promoters for use in Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J.
Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
[0329] Methods of transforming Pichia for expression of foreign genes are taught in, for example, Cregg et al. (1993), and various Phillips patents (e.g. US 4 857 467), and Pichia expression kits are commercially available from Invitrogen By, Leek, Netherlands, and Invitrogen Corp., San Diego, California.
Suitable promoters include AOXI and A0X2. Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1; whilst EP 361 991, Fleer et al. (1991) and other-publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces app., a suitable promoter being PGKI.
[0330] The transcription termination signal is preferably the 3' flanking sequence of a eukaryotic gene which contains proper = signals for transcription termination and polyadenylation. Suitable 3' flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter.
Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
[0331] The desired albumin fusion protein may be initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen.
Leaders useful in yeast include any of the following:
a) the MPIF-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMINTALGSQA (SEQ NO:2132) b) the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ NO:1054) c) the pre-pro region of the HSA signal sequence (e.g., MKWITISLLFLFSSAYSRGVFRR, SEQ ID NO:1176) d) the pre region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYS, SEQ
ED NO:1177) or variants thereof, such as, for example, MKWVSFISLLFLFSSAYS, (SEQ ID NO:1168) e) the invertase signal sequence (e.g., MLLQAFLFLLAGFAAKISA, SEQ II) NO:1108) 0 the yeast mating factor alpha signal sequence (e.g., AVLPFSNSTNNGLLFINTTIASIAAKEEGVSLEKR, SEQ ID NO:1109 or MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDV
AVLPFSNSTNNGLLFINTTIASIAAKEEGVSLDKR, SEQ ID NO:1109) g) K lactis killer toxin leader sequence h) a hybrid signal sequence (e.g., MKWVSFISLLFLFSSAYSRSLEKR, SEQ ID
NO:1110) i) an HSA/MFa-1 hybrid signal sequence (also known as HSA/kex2) (e.g., MKWVSFISLLFLFSSAYSRSLDICR, SEQ ID NO:1111) j) a K lactis killer/ MFa-1 fusion leader sequence (e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO:1169) k) the Immunoglobulin Ig signal sequence (e.g., MGWSCIILFLVATATGVHS, SEQ
ID NO:1095) 1) the Fibulin B precursor signal sequence (e.g., MERAAPSRRVPLPLLLLGGLALLAAGVDA, SEQ ID NO:1096) =
= m) the clusterin precursor signal sequence (e.g., MMKTLLLFVGLLLTWESGQVLG, SEQ ID NO:1097) n) the insulin-like growth factor-binding protein 4 signal sequence (e.g., MLPLCLVAALLLAAGPGPSLG, SEQ ID NO:1098) o) variants of the pre-pro-region of the HSA signal sequence such as, for example, MKWVSFISLLFLFSSAYSRGVFRR (SEQ ID N6:1167), MKWVTFISLLFLFAGVLG (SEQ ID NO:1099), MKWVTFISLLFLFSGVLG (SEQ ID NO:1100), MKWVTFISLLFLFGGVLG (SEQ ID NO:1101), Modified HSA leader HSA #64 MKWVTFISLLFLFAGVSG (SEQ ID NO:2133);
Modified HSA leader HSA #66 MKWVTFISLLFLFGGVSG (SEQ ID NO:2134);
Modified HSA (A14) leader ¨
MKWVTFISLLFLFAGVSG (SEQ ID NO: 1102);
Modified HSA (S14) leader (also known as modified HSA #65) ¨
MKWVTFISLLFLFSGVSG (SEQ ID NO:1103), Modified HSA (G14) leader ¨

MKWVTFISLLFLFGGVSG (SEQ ID NO:1104), or MKWVTFISLLFLFGGVLGDLHKS (SEQ ID NO:1105) p) a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID
NO:1055) q) acid phosphatase (P1105) leader (e.g., MFKSVVYSILAASLANA SEQ ID
NO:2135) r) the pre-sequence of MFoz-1 s) the pre-sequence of 0 glucanase (BGL2) t) killer toxin leader u) the presequence of killer toxin v) k. lactis killer toxin prepro (29 amino acids; 16 amino acids of pre and 13 amino acids of pro) MNIFYIFLFLLSFVQGLEHTHRRGSLDKR (SEQ ID NO:2136) w) S. diastaticus glucoarnylase Ii secretion leader sequence x) S. carlsbergensis a-galactosidase (MEL1) secretion leader sequence y) Candida glucoarnylase leader sequence z) The hybrid leaders disclosed in EP-A-387 319 aa) the gp67 signal sequence (in conjunction with baculoviral expression systems) (e.g., amino acids 1-19 of GenBank Accession Number AAA72759) or bb) the natural leader of the therapeutic protein X;
cc) S. cerevisiae invertase (SUC2) leader, as disclosed in JP 62-096086 (granted as 911036516); or dd) Inulinase ¨ MKLAYSLLLPLAGVSASVINYICR (SEQ ID NO:2137).
ee) A modified TA57 propeptide leader variant #1 ¨
MICLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTN
SGGLDVVGLISMAKR (SEQ ID NO:2128) if) A modified TA57 propeptide leader variant #2 ¨
MKLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQIN
SGGLDVVGLLSMAEEGEPKR (SEQ ID NO:2129) Additional Methods of Recombinant and Synthetic Production of Albumin Fusion Proteins [0332] The present invention also relates to vectors containing a polynucleotide encoding an albumin fusion protein of the present invention, host cells, and the production of albumin fusion proteins by synthetic and recombinant techniques. The vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
[0333] The polynucleotides encoding albumin fusion proteins of the invention may be joined to a vector containing a selectable marker for propagation in a host.
Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
103341 The polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral L'TRs, to name a few. Other suitable promoters will be known to the skilled artisan. The expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
[0335] As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E.
coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No. 201178)); insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
[0336] Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pN8A, pNH16a, pNH18A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pK1(223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, p0G44, pXT1 and pSG available from , Stratagene; and pSVIC3, pBPV, pMSG and pSVL available from Pharmacia.
Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PA0815 (all available from Invitrogen, Carlbad, CA).
Other suitable vectors will be readily apparent to the skilled artisan.
103371 In one embodiment, polynucleotides encoding an albumin fusion protein of the invention may be fused to signal sequences which will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
For example, in E. colt, one may wish to direct the expression of the protein to the periplasmic space.
Examples of signal sequences or proteins (or fragments thereof) to which the albumin fusion proteins of the invention may be fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB
signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase. Several vectors are commercially available for the construction of fusion proteins which will direct the localization of a protein, such as the pMAL series of vectors (particularly the pMAL-p series) available from New England Biolabs.
In a specific embodiment, polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Patent Nos. 5,576,195 and 5,846,818.
103381 Examples of signal peptides that may be fused to an albumin fusion protein of the invention in order to direct its secretion in mammalian cells include, but are not limited to:
a) the MPIF-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMLVTALGSQA (SEQ ID NO:2132) b) the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID NO:1054) c) the pre-pro region of the HSA signal sequence (e.g., MK'WVTFISLLFLFSSAYSRGVFRIt, SEQ ID NO:1176) d) the pre region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYS, SEQ
ID NO:1177) or variants thereof, such as, for example, MKWVSFISLLFLFSSAYS, (SEQ ID NO:1168) e) the invertase signal sequence (e.g., MLLQAFLFLLAGFAAKISA, SEQ ID
NO:1108) 0 the yeast mating factor alpha signal sequence (e.g., MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDVAVL
PFSNSTNNGLLFINTTIASIAAKEEGVSLEKR, SEQ ID NO:1109 or MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDVAVL
PFSNSTNNGLLFINTTIASIAAKEEGVSLDKR, SEQ ID NO:1109) g) K. lactis killer toxin leader sequence h) a hybrid signal sequence (e.g., MKWVSFISLLFLFSSAYSRSLEKR, SEQ ID
NO:1110) i) an HSA/MFa-1 hybrid signal sequence (also known as HSA/kex2) (e.g., MKWVSFISLLFLFSSAYSRSLDKR, SEQ ID NO:1111) j) a K lactis killer/ MFa-1 fusion leader sequence (e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO:1169) k) the Itnmunoglobulin Ig signal sequence (e.g., MGWSCIILFLVATATGVHS, SEQ
ID NO:1095) 1) the Fibulin B precursor signal sequence (e.g., MERAAPSRRVPLPILLLGGLALLAAGVDA, SEQ ID NO:1096) m) the clusterin precursor signal sequence (e.g., MMKTLLLFVGLLLTWESGQVLG, SEQ ID NO:1097) n) the insulin-like growth factor-binding protein 4 signal sequence (e.g., MLPLCLVAALLLAAGPGPSLG, SEQ ID NO:1098) o) variants of the pre-pro-region of the HSA signal sequence such as, for example, MKWVSFISLLFLFSSAYSRGVFRR (SEQ ID NO:1167), MKWVTFISLLFLFAGVLG (SEQ ID NO:1099), MK'WVTFISLLFLFSGVLG (SEQ ID NO:1100), MKWVTFISLLFLFGGVLG (SEQ ID NO:1101), Modified HSA leader HSA #64 MKWVTFISLLFLFAGVSG (SEQ ID NO:2133);
Modified HSA leader HSA #66 MKWVTFISLLFLFGGVSG (SEQ NO:2134);
Modified HSA (A14) leader ¨
MKWVTFISLLFLFAGVSG (SEQ ID NO: 1102);

Modified HSA (S14) leader (also known as modified HSA #65) ¨
MKWVTFISLLFLFSGVSG (SEQ ID NO:1103), Modified HSA (G14) leader ¨
MKWVTFISLLFLFGGVSG (SEQ ID NO:1104), or MKWVTFISLLFLFGGVLGDLHKS (SEQ ID NO:1105) p) a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ
NO:1055) q) acid phosphatase (PH05) leader (e.g., MFKSV'VYSILAASLANA SEQ ID
NO:2135) r) the pre-sequence of MFoz-1 s) the pre-sequence of 0 glucanase (BGL2) t) killer toxin leader u) the presequence of killer toxin v) k. lactis killer toxin prepro (29 amino acids; 16 amino acids of pre and 13 amino acids of pro) MNIFYIFLFLLSFVQGLEHTFIRRGSLDICR (SEQ ID NO:2136) w) S. diastaticus glucoarnylase 11 secretion leader sequence x) S. carlsbergensis a-galactosidase (MEL1) secretion leader sequence y) Candida glucoarnylase leader sequence z) The hybrid leaders disclosed in EP-A-387 319 aa) the gp67 signal sequence (in conjunction with baculoviral expression systems) (e.g., amino acids 1-19 of GenBank Accession Number AAA72759) or bb) the natural leader of the therapeutic protein X;
cc) S. cerevisiae invertase (SUC2) leader, as disclosed in JP 62-096086 (granted as 911036516); or dd) Inulinase ¨ MKLAYSLLLPLAGVSASVINYKR (SEQ ID NO:2137).
cc) A modified TA57 propeptide leader variant #1 ¨
MKLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTNSGG
LDVVGLISMAKR (SEQ ID NO:2128) if) A modified TA57 propeptide leader variant #2 ¨
MKLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTNSG
GLDVVGLISMAEEGEPKR (SEQ ID NO:2129) [0339] Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications: W087/04462;
W086/05807; W089/01036; W089/10404; and W091/06657.
Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologics, Inc. (Portsmouth, NH).
Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 1 1:1 (1995), [03401 The present invention also relates to host cells containing the above-described vector constructs described herein, and additionally encompasses host cells containing nucleotide sequences of the invention that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art. The host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. A host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled. Furthermore, different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
[0341] Introduction of the nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Bask Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
103421 In addition to encompassing host cells containing the vector constructs discussed herein, the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Therapeutic protein may be replaced with an albumin fusion protein corresponding to the Therapeutic protein), and/or to include genetic material (e.g., heterologous polynucleotide sequences such as for example, an albumin fusion protein of the invention corresponding to the Therapeutic protein may be included). The genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
[03431 In addition, techniques known in the art may be used to operably associate heterologous polynucleotides (e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof) and/or heterologous control regions (e.g., promoter and/or enhancer) with endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., US Patent Number 5,641,670, issued June 24, 1997;
International Publication Number WO 96/29411; International Publication Number WO
94/12650; Koller et al., Proc. Natl. Acad. ScL USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989)).
[03441 Albumin fusion proteins of the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, hydrophobic charge interaction chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification.
[03451 In preferred embodiments the albumin fusion proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharosiem, DEAR sepharoZ poros HQ, poros DEAR, Toyopearl Q, Toyopearl QAE, Toyopearl DEAR, Resource/Source Q and DEAR, Fractogel Q and DEAR
columns.
[03461 In specific embodiments the albumin fusion proteins of the invention are TM
purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharosTeA, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM columns and their equivalents and comparables.
[03471 In specific embodiments the albumin fusion proteins of the invention are purified using Hydrophobic Interaction Chromatography including, but not limited to, Phenyl, Butyl, Methyl, Octyl, Hexyl-sepharosTem, poros Phenyl, Butyl, Methyl, Octyl, Hexyl, Toyopearl Phenyl, Butyl, Methyl, Octyl, Hexyl Resource/Source Phenyl, Butyl, Methyl, Octyl, Hexyl, Fractogel Phenyl, Butyl, Methyl, Octyl, Hexyl columns and their equivalents and comparables.
[0348) In specific embodiments the albumin fusion proteins of the invention are purified using Size Exclusion Chromatography including, but not limited to, sepharog S100, S200, S300, superdex resin columns and their equivalents and comparables.
103491 In specific embodiments the albumin fusion proteins of the invention are purified using Affinity Chromatography including, but not limited to, Mimetic Dye affinity, peptide affinity and antibody affinity columns that are-selective for either the HSA or the "fusion target" molecules.
[0350] In preferred embodiments albumin fusion proteins of the invention are purified using one or more Chromatography methods listed above. In other preferred embodiments, albumin fusion proteins of the invention are purified using one or more of the following Chromatography columns, Q sepharoaA FF column, SP SepharosTe4FF column, Q
SepharosTem High Performance Column, Blue SepharoieFF column , Blue Column, Phenyl Sepharde"FF
column, DEAE SepharosTemFF, or Methyl Column.
103511 Additionally, albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO 00/44772.
One of skill in the art could easily modify the process described therein for use in the purification of albumin fusion proteins of the invention.
[0352] Albumin fusion proteins of the present invention may be recovered from:
products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eulcaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, albumin fusion proteins of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
Thus, it is well known in the art that the N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked.
[0353] In one embodiment, the yeast Pichia pastoris is used to express albumin fusion proteins of the invention in a eukaryotic system. Pichia pastoris is a methylotrophic yeast which can metabolize methanol as its sole carbon source. A main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde using 02.
This reaction is catalyzed by the enzyme alcohol oxidase. In order to metabolize methanol as its sole carbon source, Pichia pastoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for 02. Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol oxidase genes (A0X1) is highly active. In the presence of methanol, alcohol oxidase produced from the A0X1 gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris. See Ellis, S.B., et al., Ma Cell. Biol. 5:1111-21(1985); Koutz, P.J, et al., Yeast 5:167-77 (1989); Tschopp, J.F., et al., Nucl. Acids Res.
15:3859-76 (1987).
Thus, a heterologous coding sequence, such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the A0X1 regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
[0354] In one example, the plasmid vector pPIC9K is used to express DNA encoding an albumin fusion protein of the invention, as set forth herein, in a Pichea yeast system essentially as described in "Pichia Protocols: Methods in Molecular Biology,"
D.R. Higgins and J. Cregg, eds. The Humana Press, Totowa, NJ, 1998. This expression vector allows expression and secretion of a polypeptide of the invention by virtue of the strong ACM
promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.
103551 Many other yeast vectors could be used in place of pPIC9K, such as, pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, and PA0815, as one skilled in the art would readily appreciate, as long as the proposed expression construct provides appropriately located signals for transcription, =
translation, secretion (if desired), and the like, including an in-frame AUG
as required.
[0356] In another embodiment, high-level expression of a heterologous coding sequence, such as, for example, a polynucleotide encoding an albumin fusion protein of the present invention, may be achieved by cloning the heterologous polynucleotide of the invention into an expression vector such as, for example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of methanol.
[0357] In addition, albumin fusion proteins of the invention can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman & Co., N.Y., and Hunkapiller et al., Nature, 310:105-111 (1984)). For example, a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer. Furthermore, if desired, nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence. Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-atninobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D
(dextrorotary) or L
(levorotary).
[0358] The invention encompasses albumin fusion proteins of the present invention which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH4; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
[0359] Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or 0-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or 0-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression. The albumin fusion proteins may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
(03601 Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine (121L 123L 125L 13j), carbon ('4C),sulfur ("S), tritium (3H), indium (ttthi Il2j, 113min, 115mIn), technetium (99Tc,99"1c), thallium (201Ti), gallium (68Ga, 67Ga), palladium (1 3Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F), 153Sm, 177Lu, 159Gd, 149Pm, '?La, 1751r1), 166H0, 90y, 47sc, 6Re, 188Re, 142- , pr 105Rh, and 97Ru.
103611 In specific embodiments, albumin fusion proteins of the present invention or fragments or variants thereof are attached to macrocyclic chelators that associate with radiometal ions, including but not limited to, 177Lu, 9 Y, I66Ho, and 153Sm, to polypeptides. In a preferred embodiment, the radiometal ion associated with the macrocyclic chelators is 1111n.
In another preferred embodiment, the radiometal ion associated with the macrocyclic chelator is 9 Y. In specific embodiments, the macrocyclic chelator is 1,4,7,10-tetraazacyclododecane-N,M,N',Nm-tetraacetic acid (DOTA). In other specific embodiments, DOTA is attached to an antibody of the invention or fragment thereof via linker molecule.
Examples of linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art - see, for example, DeNardo et al., Clin Cancer Res.
4(10)2483-90 (1998);
Peterson et al., Bioconjug. Chem. 10(4):553-7 (1999); and Zimmerman et al, Nucl. Med.
Biol. 26(8):943-50 (1999), 103621 As mentioned, the albumin fusion proteins of the invention may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. (See, for instance, PROTEINS - STRUCTURE AND MOLECULAR
PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993);
POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth.
Enzymol. 182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48-62 (1992)).

Albumin fusion proteins of the invention and antibodies that bind a Therapeutic protein or fragments or variants thereof can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci.
USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the "HA"
tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the "flag" tag.
[0364]
Further, an albumin fusion protein of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi. A
cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
[0365] The conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, I3-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., INF-alpha, TNF-beta, AIM I
(See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fos Ligand (Takahashi et al., Int. Immunol., 6:1567-1574 (1994)), VEGI
(See, International Publication No. WO 99/23105), a thrombotic agent or an anti- angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("EL-1"), interleukin-2 ("IL-2"), interleuldn-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors. Techniques for conjugating such therapeutic moiety to proteins (e.g., albumin fusion proteins) are well known in the art.
[0366]
Albumin fusion proteins may also be attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
[0367]
Albumin fusion proteins, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.

[0368] In embodiments where the albumin fusion protein of the invention comprises only the VH domain of an antibody that binds a Therapeutic protein, it may be necessary and/or desirable to coexpress the fusion protein with the VL domain of the same antibody that binds a Therapeutic protein, such that the VH-albumin fusion protein and VL
protein will associate (either covalently or non-covalently) post-translationally.
[0369] In embodiments where the albumin fusion protein of the invention comprises only the VL domain of an antibody that binds a Therapeutic protein, it may be necessary and/or desirable to coexpress the fusion protein with the VH domain of the same antibody that binds a Therapeutic protein, such that the VL-albumin fusion protein and VII protein will associate (either covalently or non-covalently) post-translationally.
[0370] Some Therapeutic antibodies are bispecific antibodies, meaning the antibody that binds a Therapeutic protein is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. In order to create an albumin fusion protein corresponding to that Therapeutic protein, it is possible to create an albumin fusion protein which has an scFv fragment fused to both the N- and C- terminus of the albumin protein moiety. More particularly, the scFv fused to the N-terminus of albumin would correspond to one of the heavy/light (VHNL) pairs of the original antibody that binds a Therapeutic protein and the scFv fused to the C-terminus of albumin would correspond to the other heavy/light (VH/VL) pair of the original antibody that binds a Therapeutic protein.
[0371] Also provided by the invention are chemically modified derivatives of the albumin fusion proteins of the invention which may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immtmogenicity (see U.S. Patent No. 4,179,337). The chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like. The albumin fusion proteins may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
[0372] The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about" indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing. Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a Therapeutic protein or analog). For example, the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000,95,000, or 100,000 lcDa.
[0373] As noted above, the polyethylene glycol may have a branched structure.
Branched polyethylene glycols are described, for example, in U.S. Patent No.
5,643,575;
Morpurgo et al., App!. Biochem. Biotechnol. 56:59-72 (1996); Vorobjev et al., Nucleosides Nucleotides /8:2745-2750 (1999); and Caliceti et al., Bioconjug. Chem. 10:638-646 (1999).
[0374] The polyethylene glycol molecules (or other chemical moieties) should be attached to the protein with consideration of effects on fimctional or antigenic domains of the protein. There are a number of attachment methods available to those skilled in the art, such as, for example, the method disclosed in EP 0 401 384 (coupling PEG to G-CSF), see also Malik et al., Exp. Hematol. 20:1028-1035 (1992), reporting pegylation of GM-CSF using tresyl chloride. For example, polyethylene glycol may be covalently bound through amino acid residues via reactive group, such as a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound. The amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue. Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules. Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
[03751 As suggested above, polyethylene glycol may be attached .to proteins via linkage to any of a number of amino acid residues. For example, polyethylene glycol can be linked to proteins via covalent bonds to .lysine, histidine, aspartic acid, glutamic acid, or cysteine residues. One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof) of the protein.
103761 One may specifically desire proteins chemically modified at the N-terminus.
Using polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein. The method of obtaining the N-terminally pegylated preparation (i.e., separating this moiety from other monopegylated moieties if necessary) may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules. Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus .with a carbonyl group containing polymer is achieved.
103771 As indicated above, pegylation of the albumin fusion proteins of the invention may be accomplished by any number of means. For example, polyethylene glycol may be attached to the albumin fusion protein either directly or by an intervening linker. Linkerless systems for attaching polyethylene glycol to proteins are described in Delgado et al., Crit.
Rev. Thera. Drug Carrier Sys. 9:249-304 (1992); Francis et al., Intern. J. of Hematol. 68:1-18 (1998); U.S. Patent No. 4,002,531; U.S. Patent No. 5,349,052; WO 95/06058; and WO 98/32466.
103781 One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening lipker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (CISO2CH2CF3). Upon reaction of protein with tresylated MPEG, polyethylene glycol is directly attached to amine groups of the protein. Thus, the invention includes protein-polyethylene glycol conjugates produced by reacting proteins of the invention with a polyethylene glycol molecule having a 2,2,2-trifluoreothane sulphonyl group.
103791 Polyethylene glycol can also be attached to proteins using a number of different intervening linkers. For example, U.S. Patent No. 5,612,460, discloses urethane linkers for connecting polyethylene glycol to proteins. Protein-polyethylene glycol conjugates wherein the polyethylene glycol is attached to the protein by a linker can also be produced by reaction of proteins with compounds such as MPEG-succinimidylsuceinate, MPEG activated with 1,11-carbonyldiimidazole, MPEG-2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-succinate derivatives. A number of additional polyethylene glycol derivatives and reaction chemistries for attaching polyethylene glycol to proteins are described in International Publication No. WO 98/32466.
Pegylated protein products produced using the reaction chemistries set out herein are included within the scope of the invention.
103801 The number of polyethylene glycol moieties attached to each albumin fusion protein of the invention (i.e., the degree of substitution) may also vary. For example, the pegylated proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 17, 20, or more polyethylene glycol molecules. Similarly, the average degree of substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-11, 10-12, 11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol moieties per protein molecule. Methods for determining the degree of substitution are discussed, for example, in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992).
103811 The polypeptides of the invention can be recovered and purified from chemical synthesis and recombinant cell cultures by standard methods which include, but are not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
103821 The presence and quantity of albumin fusion proteins of the invention may be determined using ELISA, a well known immunoassay known in the art. In one ELISA
protocol that would be useful for detecting/quantifying albumin fusion proteins of the invention, comprises the steps of coating an ELISA plate with an anti-human serum albumin antibody, blocking the plate to prevent non-specific binding, washing the ELISA plate, adding a solution containing the albumin fusion protein of the invention (at one or more different concentrations), adding a secondary anti-Therapeutic protein specific antibody coupled to a detectable label (as described herein or otherwise known in the art), and detecting the presence of the secondary antibody. In an alternate version of this protocol, the ELISA plate might be coated with the anti-Therapeutic protein specific antibody and the labeled secondary reagent might be the anti-human albumin specific antibody.
Uses oldie Polynucleotides [0383] Each of the polynucleotides identified herein can be used in numerous ways as reagents. The following description should be considered exemplary and utilizes known techniques.
[0384] The polynucleotides of the present invention are useful to produce the albumin fusion proteins of the invention. As described in more detail below, polynucleotides of the invention (encoding albumin fusion proteins) may be used in recombinant DNA
methods useful in genetic engineering to make cells, cell lines, or tissues that express the albumin fusion protein encoded by the polynucleotides encoding albumin fusion proteins of the invention.
[0385] Polynucleotides of the present invention are also useful in gene therapy. One goal of gene therapy is to insert a normal gene into an organism having a defective gene, in an effort to correct the genetic defect. The polynucleotides disclosed in the present invention offer a means of targeting such genetic defects in a highly accurate manner.
Another goal is to insert a new gene that was not present in the host genome, thereby producing a new trait in the host cell. Additional non-limiting examples of gene therapy methods encompassed by the present invention are more thoroughly described elsewhere herein (see, e.g., the sections labeled "Gene Therapy", and Examples 63 and 64).
Uses of the Polypeptides [0386] Each of the polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
[0387] Albumin fusion proteins of the invention are useful to provide immunological probes for differential identification of the tissue(s) (e.g., immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem.
29:577-580 (1981)) or cell type(s) (e.g., immunocytochemistry assays).
[0388] Albumin fusion proteins can be used to assay levels of polypeptides in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell. Biol.
105:3087-3096 (1987)). Other methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable assay labels are known in the art and include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (1311, 1251, 1231, 121=s, 1) carbon (14C), sulfur (35S), tritium (3H), indium (115m ii3m1n, 112= , .tn "In), and technetium (99Tc, 99mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (1 3Pd), molybdenum ("Mo), xenon (133Xe), fluorine (18F), , 153-m S 177LU, 159Gd, 149PM, 140La, 175yb, 166H0, 90y, 47sc, 186Re, 188Re, 142pr, 105=,.it.n , Ru; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0389]
Albumin fusion proteins of the invention can also be detected in vivo by imaging. Labels or markers for in vivo imaging of protein include those detectable by X-radiography, nuclear magnetic resonance (NMR) or electron spin relaxtion (ESR). For X-radiography, suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject. Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients given to a cell line expressing the albumin fusion protein of the invention.
[0390] An albumin fusion protein which has been labeled with an appropriate , 99M, , detectable imaging moiety, such as a radioisotope (for example, 131/ 112-Tc, (1311 125j 123/, 121=µ, t.) carbon (14C), sulfur (35S), tritium (3H), indium (115mIn, tomin,lI2 "In), and technetium (99Tc, 99mTc), thallium (201Ti), gallium (68Ga, 6 -7Ga), palladium (1 3Pd), , molybdenum (99Mo), xenon (133Xe), fluorine (18F, 153Sm, mLu, 159Gd, 149pm, 140La, 175yb 166H0, 90y, 47sc, 186Re, 188Re, 142pr, 105=..tcn. , or Ru), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for immune system disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. The labeled albumin fusion protein will then preferentially accumulate at locations in the body (e.g., organs, cells, extracellular spaces or matrices) where one or more receptors, ligands or substrates (corresponding to that of the Therapeutic protein used to make the albumin fusion protein of the invention) are located. Alternatively, in the case where the albumin fusion protein comprises at least a fragment or variant of a Therapeutic antibody, the labeled albumin fusion protein will then preferentially accumulate at the locations in the body (e.g., organs, cells, extracellular spaces or matrices) where the polypeptides/epitopes corresponding to those bound by the Therapeutic antibody (used to make the albumin fusion protein of the invention) are located. In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments"
(Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A.
Rhodes, eds., Masson Publishing Inc. (1982)). The protocols described therein could easily be modified by one of skill in the art for use with the albumin fusion proteins of the invention.
[0391] In one embodiment, the invention provides a method for the specific delivery of albumin fusion proteins of the invention to cells by administering albumin fusion proteins of the invention (e.g., polypeptides encoded by polynucleotides encoding albumin fusion proteins of the invention and/or antibodies) that are associated with heterologous polypeptides or nucleic acids. In one example, the invention provides a method for delivering a Therapeutic protein into the targeted cell. - In another example, the invention provides a method for delivering a single stranded nucleic acid (e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed) into the targeted cell.
[0392] In another embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering albumin fusion proteins of the invention in association with toxins or cytotoxic prodrugs.
[0393] By "toxin" is meant one or more compounds that bind and activate endogenous cytotoxic effector systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of toxins, or any molecules or enzymes not normally present in or on the surface of a cell that under defined conditions cause the cell's death. Toxins that may be used according to the methods of the invention include, but are not limited to, radioisotopes known in the art, compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin. "Toxin" also includes a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 2I3Bi, or other radioisotopes such as, for example, mpd, 133xe, 1311, , 68¨e CI "Co, 65Zn, "Sr, "P, 35S, 9 Y, 153Sm, '"Gd, '69Y 'O 13 "Cr, "Mn, "Se, "3Sn, "Yttrium, 117Tin, 186Rhenium, 166Holmium, and 188Rhenium;
luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin. In a specific embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 90Y. In another specific embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 111In. In a further specific embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 1311.
103941 Techniques known in the art may be applied to label polypeptides of the invention. Such techniques include, but are not limited to, the use of bifunctional conjugating agents (see e.g., U.S. Patent NQS. 5,756,065; 5,714,631;
5,696,239; 5,652,361;
5,505,931; 5,489,425; 5,435,990; 5,428,139; 5,342,604; 5,274,119; 4,994,560;
and 5,808,003).
103951 The albumin fusion proteins of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans.
Such disorders include, but are not limited to, those described herein under the section heading "Biological Activities," below.
103961 Thus, the invention provides a diagnostic method of a disorder, which involves (a) assaying the expression level of a certain polypeptide in cells or body fluid of an individual using an albumin fusion protein of the invention; and (b) comparing the assayed polypeptide expression level with a standard polypeptide expression level, whereby an increase or decrease in the assayed polypeptide expression level compared to the standard expression level is indicative of a disorder. With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.

[0397] Moreover, albumin fusion proteins of the present invention can be used to treat or prevent diseases or conditions such as, for example, neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions. For example, patients can be administered a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins), to inhibit the activity of a polypeptide (e.g., an oncogene or tumor supressor), to activate the activity of a polypeptide (e.g., by binding to a receptor), to reduce the activity of a membrane bound receptor by competing with it for free ligand (e.g., soluble TNF receptors used in reducing inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of the immune response to proliferative cells or tissues).
[0398] In particular, albumin fusion proteins comprising of at least a fragment or variant of a Therapeutic antibody can also be used to treat disease (as described supra, and elsewhere herein). For example, administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can bind, and/or neutralize the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, and/or reduce overproduction of the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds.
Similarly, administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can activate the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, by binding to the polypeptide bound to a membrane (receptor).
[0399] At the very least, the albumin fusion proteins of the invention of the present invention can be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art.
Albumin fusion proteins of the invention can also be used to raise antibodies, which in turn may be used to measure protein expression of the Therapeutic protein, albumin protein, and/or the albumin fusion protein of the invention from a recombinant cell, as a way of assessing transformation of the host cell, or in a biological sample. Moreover, the albumin fusion proteins of the present invention can be used to test the biological activities described herein.

Diagnostic Assays [0400] The compounds of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans. Such disorders include, but are not limited to, those described for each Therapeutic protein in the corresponding row of Table 1 and herein under the section headings "Immune Activity,"
"Blood Related Disorders," "Hyperproliferative Disorders," "Renal Disorders,"
"Cardiovascular Disorders," "Respiratory Disorders," "Anti-Angiogenesis Activity,"
"Diseases at the Cellular Level," "Wound Healing and Epithelial Cell Proliferation," "Neural Activity and Neurological Diseases," "Endocrine Disorders," "Reproductive System Disorders," "Infectious Disease," "Regeneration," and/or "Gastrointestinal Disorders," infra.
[0401] For a number of disorders, substantially altered (increased or decreased) levels of gene expression can be detected in tissues, cells or bodily fluids (e.g., sera, plasma, urine, semen, synovial fluid or spinal fluid) taken from an individual having such a disorder, relative to a "standard" gene expression level, that is, the expression level in tissues or bodily fluids from an individual not having the disorder. Thus, the invention provides a diagnostic method useful during diagnosis of a disorder, which involves measuring the expression level .
of the gene encoding a polypeptide in tissues, cells or body fluid from an individual and comparing the measured gene expression level with a standard gene expression level, whereby an increase or decrease in the gene expression level(s) compared to the standard is indicative of a disorder. These diagnostic assays may be performed in vivo or in vitro, such as, for example, on blood samples, biopsy tissue or autopsy tissue.
[0402] The present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome [04031 By "assaying the expression level of the gene encoding a polypeptide" is intended qualitatively or quantitatively measuring or estimating the level of a particular polypeptide (e.g. a polypeptide corresponding to a Therapeutic protein disclosed in Table 1) or the level of the mRNA encoding the polypeptide of the invention in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA
level) or relatively (e.g., by comparing to the polypeptide level or mRNA
level in a second biological sample). Preferably, the polypeptide expression level or mRNA level in the first = biological sample is measured or estimated and compared to a standard polypeptide level or mRNA level, the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having the disorder. As will be appreciated in the art, once a standard polypeptide level or mRNA level is known, it can be used repeatedly as a standard for comparison.
[0404] By "biological sample" is intended any biological sample obtained from an individual, cell line, tissue culture, or other source containing polypeptides of the invention (including portions thereof) or mRNA. As indicated, biological samples include body fluids (such as sera, plasma, urine, synovial fluid and spinal fluid) and tissue sources found to express the full length or fragments thereof of a polypeptide or mRNA. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. Where the biological sample is to include mRNA, a tissue biopsy is the preferred source.
[0405] Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynsld and Sacchi, Anal. Biochem. 162:156-159 (1987). Levels of mRNA encoding the polypeptides of the invention are then assayed using any appropriate method. These include Northern blot analysis, Si nuclease mapping, the polymerase chain reaction (PCR), reverse transcription in combination with the polymerase chain reaction (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR).
[0406] The present invention also relates to diagnostic assays such as quantitative and diagnostic assays for detecting levels of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the invention, in a biological sample (e.g., cells and tissues), including determination of normal and abnormal levels of polnieptides. Thus, for instance, a diagnostic assay in accordance with the invention for detecting abnormal expression of polypeptides that bind to, are bound by, or associate with albumin fusion proteins compared to normal control tissue samples may be used to detect the presence of tumors.
Assay techniques that can be used to determine levels of a polypeptide that bind to, are bound by, or associate with albumin fusion proteins of the present invention in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA
assays.
Assaying polypeptide levels in a biological sample can occur using any art-known method.
[0407]
Assaying polypeptide levels in a biological sample can occur using a variety of techniques. For example, polypeptide expression in tissues can be studied with classical immunohistological methods (Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., et al., J. Cell . Biol. 105:3087-3096 (1987)). Other methods useful for detecting =

polypeptide gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase, and ,, 121*s radioisotopes, such as iodine (1251 i) carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99mTc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0408) The tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the gene of interest (such as, for example, cancer). The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York).
The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be a necessary step in the assessment of cells that could be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the gene.
[04091 For example, albumin fusion proteins may be used to quantitatively or qualitatively detect the presence of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the present invention. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled albumin fusion protein coupled with light microscopic, flow cytometric, or fluorimetric detection.
[04101 In a preferred embodiment, albumin, fusion proteins comprising at least a fragment or variant of an antibody that specifically binds at least a Therapeutic protein disclosed herein (e.g., the Therapeutic proteins disclosed in Table 1) or otherwise known in the art may be used to quantitatively or qualitatively detect the presence of gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody coupled with light microscopic, flow cytomettic, or fluorimetric detection.
[0411) The albumin fusion proteins of the present invention may, additionally, be employed histologically, as in immunofluorescence, inummoelectron microscopy or non-immunological assays, for in situ detection of polypeptides that bind to, are bound by, or associate with an albumin fusion protein of the present invention. In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody or polypeptide of the present invention. The albumin fusion proteins are preferably applied by overlaying the labeled albumin fusion proteins onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the polypeptides that bind to, are bound by, or associate with albumin fusion proteins, but also its distribution in the examined tissue. Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.
[0412) Immunoassays and non-immunoassays that detect polypeptides that bind to, are bound by, or associate with albumin fusion proteins will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of binding gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.
[04131 The biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled albumin fusion protein of the invention. The solid phase support may then be washed with the buffer a second time to remove unbound antibody or polypeptide. Optionally the antibody is subsequently labeled.
The amount of bound label on solid support may then be detected by conventional means.
[0414] By "solid phase support or carrier" is intended any support capable of binding a polypeptide (e.g., an albumin fusion protein, or polypeptide that binds, is bound by, or associates with an albumin fusion protein of the invention.) Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention. The support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to a polypeptide. Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc.
Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.
[0415] The binding activity of a given lot of albumin fusion protein may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.
[0416] In addition to assaying polypeptide levels in a biological sample obtained from an individual, polypeptide can also be detected in vivo by imaging. For example, in one embodiment of the invention, albumin fusion proteins of the invention are used to image diseased or neoplastic cells.
[0417] Labels or markers for in vivo imaging of albumin fusion proteins of the invention include those detectable by X-radiography, NMR, MRI, CAT-scans or ESR. For X-radiography, suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject. Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients of a cell line (or bacterial or yeast strain) engineered.
[0418) Additionally, albumin fusion proteins of the invention whose presence can be detected, can be administered. For example, albumin fusion proteins of the invention labeled with a radio-opaque or other appropriate compound can be administered and visualized in vivo, as discussed, above for labeled antibodies. Further, such polypeptides can be utilized for in vitro diagnostic procedures.
[0419] A polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, 1311, 1121-m , 9T
c), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for a disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99"1Tc.
The labeled albumin fusion protein will then preferentially accumulate at the locations in the body which contain a polypeptide or other substance that binds to, is bound by or associates with an albumin fusion protein of the present invention. In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments" (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W.
Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
[0420] One of the ways in which an albumin fusion protein of the present invention can be detectably labeled is by linking the same to a reporter enzyme and using the linked product in an enzyme immunoassay (EIA) (Voller, A., "The Enzyme Linked Irrununosorbent Assay (ELISA)", 1978, Diagnostic Horizons 2:1-7, Microbiological Associates Quarterly Publication, Walkersville, MD); Voller et al., J. Clin. Pathol. 31:507-520 (1978); Butler, J.E., Meth. Enzymol. 73:482-523 (1981); Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC
Press, Boca Raton, FL,; Ishikawa, E. et al., (eds.), 1981, Enzyme Immunoassay, Kgalcu Shoin, Tokyo). The reporter enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Reporter enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose coddtise, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. Additionally, the detection can be accomplished by colorimetric methods- which employ a chromogenic substrate for the reporter enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
[0421] Albumin fusion proteins may also be radiolabelled and used in any of a variety of other immunoassays. For example, by radioactively labeling the albumin fusion proteins, it is possible to the use the albumin fusion proteins in a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioinununoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986).
The radioactive isotope can be detected by means including, but not limited to, a gamma counter, a scintillation counter, or autoradiography.
[0422] Additionally, chelator molecules, are known in the art and can be used to label the Albumin fusion proteins. Chelator molecules may be attached Albumin fusion proteins of the invention to facilitate labeling said protein with metal ions including radionuclides or fluorescent labels. For example, see Subramanian, R. and Meares, C.F., "Bifunctional Chelating Agents for Radiometal-labeled monoclonal Antibodies," in Cancer Imaging with Radiolabeled Antibodies (D. M. Goldenberg, Ed.) Kluwer Academic Publications, Boston;
Saji, H., "Targeted delivery of radiolabeled imaging and therapeutic agents:
bifunctional radiopharmaceuticals." Crit. Rev. Ther. Drug Carrier Syst. /6:209-244 (1999);
Srivastava S.C. and Mease R.C., "Progress in research on ligands, nuclides and techniques for labeling monoclonal antibodies." Int. J. Rad. App!. Instrum. B /8:589-603 (1991); and Liu, S. and Edwards, D.S., "Bifunctional chelators for therapeutic lanthanide radiopharmaceuticals."
Bioconjug. Chem. /2:7-34 (2001). Any chelator which can be covalently bound to said Albumin fusion proteins may be used according to the present invention. The chelator may further comprise a linker moiety that connects the chelating moiety to the Albumin fusion protein.
[0423] In one embodiment, the Albumin fusion protein of the invention are attached to an acyclic chelator such as diethylene triamine-N,N,N',N",N"-pentaacetic acid (DPTA), analogues of DPTA, and derivatives of DPTA. As non-limiting examples, the chelator may be 2-(p-isothiocyanatobenzy1)-6- methyldiethylenetriaminepentaacetic acid (1B4M-DPTA, also known as MX-DTPA), 2-methyl-6-(rho-nitrobenzy1)-1,4,7- triazaheptane-N,N,N,N",N"-pentaacetic acid (nitro-1B4M-DTPA or nitro-MX-DTPA); 2-(p-isothiocyanatobenzy1)-cyclohexyldiethylenetriaminepentaacetic acid (CHX-DTPA), or N42-amino-3-(rho-nitrophenyl)propylFtrans-cyclohexane-1,2-diamine-N,N,N"-pentaacetic acid (nitro-CHX-A-DTPA).
[0424] In another embodiment, the Albumin fusion protein of the invention are attached to an acyclic terpyridine chelator such as 6,6"-bisHN,N,N",N"-tetra(carboxymethyl)arnino]methyli-4'-(3-amino-4-methoxypheny1)-2,2%6',2 "-terpyridine (TMT-amine).
[0425] In specific embodiments, the macrocyclic chelator which is attached to the the Albumin fusion protein of the invention is 1,4,7,10-tetraazacyclododecane-N,N,N",N"-tetraacetic acid (DOTA). In other specific embodiments, the DOTA is attached to the the Albumin fusion protein of the invention via a linker molecule. Examples of linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art -see, for example, DeNardo et al., Clin. Cancer Res. 4(10):2483-90, 1998; Peterson et al., Bioconjug.
Chem. 10(4):553-7, 1999; and Zimmerman et al., NucL Med. Biol. 26(8):943-50, 1999.
In addition, U.S. Patents 5,652,361 and 5,756,065, which disclose chelating agents that may be conjugated to antibodies, and methods for making and using them.
Though U.S. Patents 5,652,361 and 5,756,065 focus on conjugating chelating agents to antibodies, one skilled in the art could readily adapt the method disclosed therein in order to conjugate chelating agents to other polypeptides.

[0426] Bifunctional chelators based on macrocyclic ligands in which conjugation is via an activated arm, or functional group, attached to the carbon backbone of the ligand can be employed as described by M. Moi et al., J. Amer. Chem. Soc. 49:2639 (1989) (2-p-nitrobenzy1-1,4,7,10-tetraazacyclododecane-N,N',N",N'"-tetraacetic acid); S.
V. Deshpande et al., J. NucL Med. 31:473 (1990); G. Ruser et al., Bioconj. Chem. 1:345 (1990);
C. J. Broan et al., J. C. S. Chem. Comm. 23:1739 (1990); and C. J. Anderson et al., J. Nud Med. 36:850 (1995).
[0427] In one embodiment, a macrocyclic chelator, such as polyazamacrocyclic chelators, optionally containing one or more carboxy, amino, hydroxamate, phosphonate, or phosphate groups, are attached to the Albumin fusion protein of the invention.
In another embodiment, the chelator is a chelator selected from the group consisting of DOTA, analogues of DOTA, and derivatives of DOTA.
[0428] In one embodiment, suitable chelator molecules that may be attached to the the Albumin fusion protein of the invention include DOXA (1-oxa-4,7,10-triazacyclododecanetriacetic acid), NOTA (1,4,7-triazacyclononanetriacetic acid), TETA
(1,4,8,11-tetraazacyclotetradecanetetraacetic acid), and THT (4'-(3-amino-4-methoxy-pheny1)-6,6"-bis(N',N-dicarboxymethyl-N-methylhydra zino)-2,2':6',2"-terpyridine), and analogs and derivatives thereof. See, e.g., Ohmono et al., J. Med. Chem. 35:

(1992); Kung et al., J. Nud Med. 25: 326-332 (1984); Jurisson et al., Chem.
Rev. 93:1137-1156 (1993); and U.S. Patent No. 5,367,080. Other suitable chelators include chelating agents disclosed in U.S. Patent Nos. 4,647,447; 4,687,659; 4,885,363; EP-A-71564;
W089/00557; and EP-A-232751.
[0429] In another embodiment, suitable macrocyclic carboxylic acid chelators which can be used in the present invention include 1,4,7,10-tetraancyclododecane-N,N,N",Nm-tetraacetic acid (DOTA); 1,4,8,12-tetrawacyclopentadecane-N,AP,N",N"-tetraacetic acid (15N4); 1,4,7-triazacyclononane-N,N',N"-triacetic acid (9N3) 1,5,9-triazacyclododecane-N,N,N"-triacetic acid (12N3); and 6-bromoacetamido-benzy1-1,4,8,11-tetraazacyclotetradecane- N,AP,N",/r-tetraacetic acid (BAT).
[0430] A
preferred chelator that can be attached to the Albumin Fusion protein of the invention is a-(5-isothiocyanato- 2-methoxypheny1)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid, which is also known as Me0-DOTA-NCS. A salt or ester of a-(5-isothiocyanato- 2-methoxypheny1)- 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid may also be used.

[0431] Albumin fusion proteins of the invention to which chelators such as those decribed are covalently attached may be labeled (via the coordination site of the chelator) with radionuclides that are suitable for therapeutic, diagnostic, or both therapeutic and diagnostic purposes. Examples of appropriate metals include Ag, At, Au, Bi, Cu, Ga, Ho, In, Lu, Pb, Pd, Pm, Pr, Rb, Re, Rh, Sc, Sr, Tc, Ti, Y, and Yb. Examples of the radionuclide used for diagnostic purposes are Fe, Gd, 111In, 67Ga, or 68Ga. In another embodiment, the radionuclide used for diagnostic purposes is 111In, or 67Ga. Examples of the radionuclide used for therapeutic purposes are 166/10, 165Dy, 90y, 115min, 52-Th, or - 72 r Ga. In one embodiment, the radionuclide used for diagnostic purposes is 166Ho or 90Y. Examples of the radionuclides used for both therapeutic and diagnostic purposes include 153Sm, 177Lu, 159Gd, 175Y10, or 47Sc.
In one embodiment, the radionuclide is 1"Sm, 'Lu, 175Yb, or 'Gd.
[0432] Preferred metal radionuclides include 90Y, 99mTc, 47Sc, 67-a, 51Cr, 177m5u, 67Cu, 167Tm, 97Ru, 188Re, 'Lu, 'Au, 47Sc, 67Ga, 51Cr, 177mSn, 67Cu, 167Tm, 95Ru, 188Re, 177Lu, 199An, 203pb and 141ce.
[0433] In a particular embodiment, Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with a metal ion selected from the group consisting of 90Y, "In, 166110, 215Bi, and 225Ac.
[0434] Moreover, y-emitting radionuclides, such as 99mTc, 67Ga, and 169yb have been approved or under investigation for diagnostic imaging, while 13-emitters, such as 67Cu, 111Ag, 186Re, and 90Y are useful for the applications in tumor therapy. Also other useful .
radionuclides include 7-emitters, such as 99mTc, 111In, 67Ga, and 169Y13, and 13-emitters, such as 67Cu, inAg, 186Re, 188 - --Re and 90Y as well as other radionuclides of interest such as 211At, 2t2Bi, 86Rh 105Rb, 153sm, 198Au, 149pm, 85sr, 142pr, 214pb, 109pd, 166H0, it and Sc.44 Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with the radionuclides described above.
[0435] In another embodiment, Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with paramagnetic metal ions including ions of transition and lanthanide metal, such as metals having atomic numbers of 21-29, 42, 43, 44, or 57-71, in particular ions of Cr, V, Mn, Fe, Co, Ni, Cu, La, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu. The paramagnetic metals used in compositions for magnetic resonance imaging include the elements having atomic numbers of 22 to 29, 42, 44 and 58-70.

[0436] In another embodiment, Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with fluorescent metal ions including lanthanides, in particular La, Ce, Pr, Nd, Pm, Sm, Eu (e.g., 152Eu), Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu.
[0437] In another embodiment, Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with heavy metal-containing reporters may include atoms of Mo, Bi, Si, and W.
[0438] It is also possible to label the albumin fusion proteins with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, ophthaldehyde and fluorescamine.
[0439] The albumin fusion protein can also be detectably labeled using fluorescence emitting metals such as 152Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA). =
[0440] The albumin fusion proteins can also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged albumin fusion protein is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
[0441]
Likewise, a bioluminescent compound may be used to label albumin fusion proteins of the present invention. Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.
Transgenic Organisms [0442]
Transgenic organisms that express the albumin fusion proteins of the invention are also included in the invention. Transgenic organisms are genetically modified organisms into which recombinant, exogenous or cloned genetic material has been transferred. Such genetic material is often referred to as a transgene. The nucleic acid sequence of the transgene may include one or more transcriptional regulatory sequences and other nucleic acid sequences such as introns, that may be necessary for optimal expression and secretion of the encoded protein. The transgene may be designed to direct the expression of the encoded protein in a manner that facilitates its recovery from the organism or from a product produced by the organism, e.g. from the milk, blood, urine, eggs, hair or seeds of the organism. The transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal. The transgene may be integrated either at a locus of a genome where that particular nucleic acid sequence is not otherwise normally found or at the normal locus for the transgene.
[0443] The term "germ cell line transgenic organism" refers to a transgenic organism in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability of the transgenic organism to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic information, then they too are transgenic organisms. The alteration or genetic inforrnation may be foreign to the species of organism to which the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene.
[0444] A
transgenic organism may be a transgenic animal or a transgenic plant.
Transgenic animals can be produced by a variety of different methods including transfection, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Patent No. 4,736,866; U.S. Patent No. 5,602,307;
Mullins et al. (1993) Hypertension 22(4):630-633; Brenin et al. (1997) Surg.
Oncol. 6(2)99-110; Tuan (ed.), Recombinant Gene Expression Protocols, Methods in Molecular Biology No. 62, Humana Press (1997)). The method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transformation of multiple nucleic acid molecules. Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Patent No. 5,489,743 and U.S. Patent No. 5,602,307.
[0445] A
number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Patent No.
4,736,866); express simian SV40 T-antigen (U.S. Patent No. 5,728,915); lack the expression of interferon regulatory factor 1 (1RP-1) (U.S. Patent No. 5,731,490); exhibit dopaminergic dysfunction (U.S, Patent No. 5,723,719); express at least one human gene which participates in blood pressure control (U.S. Patent No. 5,731,489); display greater similarity to the conditions existing in naturally occurring Alzheimer's disease (U.S. Patent No.
5,720,936); have a reduced capacity to mediate cellular adhesion (U.S. Patent No. 5,602,307);
possess a bovine growth hormone gene (Clutter et al. (1996) Genetics 143(4):1753-1760); or, are capable of generating a fully human antibody response (McCarthy (1997) The Lancet 349(9049):405).
[04461 While mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal species. Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g., Kim et al. (1997) Mol. Reprod. Dev. 46(4):515-526;
Houdebine (1995) Reprod. Nutr. Dev. 35(6):609-617; Petters (1994) Reprod.
Fertil. Dev.
6(5):643-645; Schnieke et al. (1997) Science 278(5346):2130-2133; and Amoah (1997) J.
Animal Science 75(2):578-585).
[04471 To direct the secretion of the transgene-encoded protein of the invention into the milk of transgenic mammals, it may be put under the control of a promoter that is preferentially activated in mammary epithelial cells. Promoters that control the genes encoding milk proteins are preferred, for example the promoter for casein, beta lactoglobulin, whey acid protein, or lactalburnin (see, e.g., DiTullio (1992) BioTechnology 10:74-77; Clark et al. (1989) BioTechnology 7:487-492; Gorton et al. (1987) BioTechnology 5:1183-1187;
and Soulier et al. (1992) FEBS Letts. 297:13). The transgenic mammals of choice would produce large volumes of milk and have long lactating periods, for example goats, cows, camels or sheep.
[0448] An albumin fusion protein of the invention can also be expressed in a transgenic plant, e.g. a plant in which the DNA transgene is inserted into the nuclear or plastidic genome. Plant transformation procedures used to introduce foreign nucleic acids into plant cells or protoplasts are known in the art. See, in general, Methods in Enzymology Vol. 153 ("Recombinant DNA Part D") 1987, Wu and Grossman Eds., Academic Press and European Patent Application EP 693554. Methods for generation of genetically engineered plants are further described in US Patent No. 5,283,184, US Patent No. 5, 482,852, and European Patent Application EP 693 554.

Pharmaceutical or Therapeutic Compositions [0449] The albumin fusion proteins of the invention or formulations thereof may be administered by any conventional method including parenteral (e.g.
subcutaneous or intramuscular) injection or intravenous infusion. The treatment may consist of a single dose or a plurality of doses over a period of time.
[0450] While it is possible for an albumin fusion protein of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers. The carrier(s) must be "acceptable" in the sense of being compatible with the albumin fusion protein and not deleterious to the recipients thereof.
Typically, the carriers will be water or saline which will be sterile and pyrogen free.
Albumin fusion proteins of the invention are particularly well suited to formulation in aqueous carriers such as sterile pyrogen free water, saline or other isotonic solutions because of their extended shelf-life in solution. For instance, pharmaceutical compositions of the invention may be formulated well in advance in aqueous form, for instance, weeks or months or longer time periods before being dispensed.
[0451] For example, formulations containing the albumin fusion protein may be prepared taking into account the extended shelf-life of the albumin fusion protein in aqueous formulations. As discussed above, the shelf-life of many of these Therapeutic proteins are markedly increased or prolonged after fusion to HA.
[0452] In instances where aerosol administration is appropriate, the albumin fusion proteins of the invention can be formulated as aerosols using standard procedures. The term "aerosol" includes any gas-borne suspended phase of an albumin fusion protein of the instant invention which is capable of being inhaled into the bronchioles or nasal passages.
Specifically, aerosol includes a gas-borne suspension of droplets of an albumin fusion protein of the instant invention, as may be produced in a metered dose inhaler or nebulizer, or in a mist sprayer. Aerosol also includes a dry powder composition of a compound of the instant invention suspended in air or other carrier gas, which may be delivered by insufflation from an inhaler device, for example. See Ganderton & Jones, Drug Delivery to the Respiratory Tract, Ellis Horwood (19 87); Gonda (1990) Critical Reviews in Therapeutic Drug Carrier Systems 6:273-313; and Raeburn et al,. (1992) Pharmacol. Toxicol. Methods 27:143-159.
[0453] The formulations of the invention are also typically non-immunogenic, in part, because of the use of the components of the albumin fusion protein being derived from the proper species. For instance, for human use, both the Therapeutic protein and albumin portions of the albumin fusion protein will typically be human. In some cases, wherein either . component is non human-derived, that component may be humanized by substitution of key amino acids so that specific epitopes appear to the human immune system to be human in nature rather than foreign.
[0454]
The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the albumin fusion protein with the carrier that constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
[0455]
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation appropriate for the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampules, vials or syringes, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders. Dosage formulations may contain the Therapeutic protein portion at a lower molar concentration or lower dosage compared to the non-fused standard formulation for the Therapeutic protein given the extended serum half-life exhibited by many of the albumin fusion proteins of the invention.
[0456] As an example, when an albumin fusion protein of the invention comprises one of the proteins listed in the "Therapeutic Protein:X" column of Table 1 as one or more of the Therapeutic protein regions, the dosage form can be calculated on the basis of the potency of the albumin fusion protein relative to the potency of hGH, while taking into account the prolonged serum half-life and shelf-life of the albumin fusion proteins compared to that of native hGH. Growth hormone is typically administered at 0.3 to 30.0 IU/kg/week, for example 0.9 to 12.0 1U/1(g/week, given in three or seven divided doses for a year or more. In an albumin fusion protein consisting of full length HA fused to full length GH, an equivalent dose in terms of units would represent a greater weight of agent but the dosage frequency can be reduced, for example to twice a week, once a week or less.
[0457] Formulations or compositions of the invention may be packaged together with, or included in a kit with, instructions or a package insert referring to the extended shelf-life of the albumin fusion protein component. For instance, such instructions or package inserts may address recommended storage conditions, such as time, temperature and light, taking into account the extended or prolonged shelf-life of the albumin fusion proteins of the invention.
Such instructions or package inserts may also address the particular advantages of the albumin fusion proteins of the inventions, such as the ease of storage for formulations that may require use in the field, outside of controlled hospital, clinic or office conditions. As described above, formulations of the invention may be in aqueous form and may be stored under less than ideal circumstances without significant loss of therapeutic activity.
[0458] Albumin fusion proteins of the invention can also be included in nutraceuticals. For instance, certain albumin fusion proteins of the invention may be administered in natural products, including milk or milk product obtained from a transgenic mammal which expresses albumin fusion protein. Such compositions can also include plant or plant products obtained from a transgenic plant which expresses the albumin fusion protein. The albumin fusion protein can also be provided in powder or tablet form, with or without other known additives, carriers, fillers and diluents. Nutraceuticals are described in Scott Hegenhart, Food Product Design, Dec. 1993.
[0459] The invention also provides methods of treatment and/or prevention of diseases or disorders (such as, for example, any one or more of the diseases or disorders disclosed herein) by administration to a subject of an effective amount of an albumin fusion protein of the invention or a polynucleotide encoding an albumin fusion protein of the invention ("albumin fusion polynucleotide") in a pharmaceutically acceptable carrier.
[0460] The albumin fusion protein and/or polynucleotide will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with the albumin fusion protein and/or polynucleotide alone), the site of delivery, the method of administration, the scheduling of administration, and other factors known to practitioners.
The "effective amount" for purposes herein is thus determined by such considerations.
[0461] As a general proposition, the total pharmaceutically effective amount of the albumin fusion protein administered parenterally per dose will be in the range of about lug/kg/day to 10 mg/kg/day of patient body weight, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most preferably for humans between about 0.01 and 1 mg/kg/day for the hormone.
If given continuously, the albumin fusion protein is typically administered at a dose rate of about 1 ug/kg/hour to about 50 ug/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump. An intravenous bag solution may also be employed. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect.
[0462] Albumin fusion proteins and/or polynucleotides can be are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray.
"Pharmaceutically acceptable carrier" refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any. The term "parenteral" as used herein refers to modes of administration which include intravenous, intramuscular, intiaperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
[0463] Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustained-release systems. Examples of sustained-release albumin fusion proteins and/or polynucleotides are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdennal patch), bucally, or as an oral or nasal spray.
"Pharmaceutically acceptable carrier" refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The term "parenteral" as used herein refers to . modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
Additional examples of sustained-release albumin fusion proteins and/or polynucleotides include suitable polymeric materials (such as, for example, semi-permeable polymer matrices in the form of shaped articles, e.g., films, or mirocapsules), suitable hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, and sparingly soluble derivatives (such as, for example, a sparingly soluble salt).
[0464] Sustained-release matrices include polylactides (U.S. Pat. No.
3,773,919, EP
58,481), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al., Biopolymers 22:547-556 (1983)), poly (2- hydroxyethyl methacrylate) (Langer et al., J.
Biomed. Mater. Res. 15:167-277 (1981), and Langer, Chem. Tech. 12:98-105 (1982)), ethylene vinyl acetate (Langer et al., Id.) or poly-D- (-)-3-hydroxybutyric acid (EP 133,988).
[0465] Sustained-release albumin fusion proteins and/or polynucleotides also include liposomally entrapped albumin fusion proteins and/or polynucleotides of the invention (see generally, Langer, Science 249:1527-1533 (1990); Treat et al., in Ltposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 317 -327 and 353-365 (1989)). Liposomes containing the albumin fusion protein and/or polynucleotide are prepared by methods known per se: DE 3,218,121; Epstein et al., Proc.
Natl. Acad. Sci. (USA) 82:3688-3692 (1985); Hwang et al., Proc. Natl. Acad.
Sci.(USA) 77:4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EP 143,949; EP 142,641;
Japanese Pat. App!. 83-118008; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324.
Ordinarily, the fiposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal Therapeutic.
[0466] In yet an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are delivered by way of a pump (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek etal., N. Engl. J. Med. 321:574 (1989)).
[0467] Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
[0468] For parenteral administration, in one embodiment, the albumin fusion protein and/or polynucleotide is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. For example, the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to the Therapeutic.
[0469] Generally, the formulations are prepared by contacting the albumin fusion protein and/or polynucleotide uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation.
Preferably the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.
[0470] The carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability. Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.

NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional volumes please contact the Canadian Patent Office.

Claims (53)

THE EMBODIMENTS OF THE INVENTION FOR WHICH AN EXCLUSIVE
PROPERTY OR PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. An albumin fusion protein comprising two tandemly oriented GLP-1 polypeptides, wherein said GLP-1 polypeptides are selected from wild-type GLP-1, GLP-1 fragments, and GLP-1 variants, fused to albumin comprising the amino acid sequence of SEQ ID NO:1038, an albumin fragment, or albumin variant thereof, wherein said albumin fragment or albumin variant increases the serum plasma half life of the GLP-1 polypeptides, and wherein said fusion protein has GLP-1 activity.
2. The albumin fusion protein of claim 1, wherein said tandemly oriented GLP-1 polypeptides are one wild-type GLP-1 polypeptide fused to one GLP-1 fragment polypeptide.
3. The albumin fusion protein of claim 1, wherein said tandemly oriented GLP-1 polypeptides are one wild type GLP-1 polypeptide fused to one GLP-1 variant polypeptide.
4. The albumin fusion protein of claim 1, wherein said tandemly oriented GLP-1 polypeptides are one GLP-1 fragment polypeptide fused to one GLP-1 variant polypeptide.
5. The albumin fusion protein of any one of claims 1 to 4, wherein said GLP-fragments or GLP-1 variants are selected from:
(a) GLP-1 (9-36);
(b) GLP-1 (7-36); and (c) GLP-1 (7-36(A8G)).
6. The albumin fusion protein of claim 5, wherein said GLP-1 fragments or GLP-1 variants are selected from two tandemly oriented GLP-1 (7-36(A8G)).
7. The albumin fusion protein of claim 1, produced from a host cell comprising a construct which expresses said albumin fusion protein, wherein said construct is selected from the following constructs:
(a) pSAC:GLP-1(7-36)x2.HSA;
(b) pSAC35:GLP-1(7-36)x2.HSA;
(c) pSAC35:GLP-1(7-36(A8G))x2.HSA;
(d) pSAC35:coGLP-1(7-36(A8G))x2.HSA;
(e) pSAC35:GLP-1(7-36(A8S)).GLP-1(7-36).HSA;
(f) pSAC35:GLP-1(7-36(A8G).GLP-1(7-36).HSA;
(g) pSAC35:KT.GLP-1(7-36(A8G))x2.HSA;
(h) pSAC35:INV.GLP-1(7-36A8G)x2.HSA;
(i) pSAC35:INV.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(j) pSAC35:DeltaKex.GLP-1(7-36A8G)x2.HSA;
(k) pSAC35:DeltaKex.GLP-1(7-36A8G).GLP-1(7-36).HSA;
(1) pSAC35:INU.GLP-1(7-36(A8G))x2.HSA;
(m) pSAC35:MAF.GLP-1(7-36(A8G))x2.HSA;
(n) pSAC35:AP.GLP-1(7-36(A8G))x2.HSA;
(o) pSAC35:MAF.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(p) pSAC35:INU.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(q) pSAC35:AP.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(r) pSAC:KT.GLP-1(7-36(A8G))x2.MSA.E25-A608; and (s) pSAC35:GLP-1(9-36).GLP-1(7-36).HSA.
8. An albumin fusion protein comprising two tandemly oriented GLP-1 polypeptides fused to an albumin comprising the amino acid sequence of SEQ ID
NO:1038, wherein said GLP-1 polypeptides comprise one of an amino acid sequence selected from:
(a) amino acids 1 to 30 of SEQ ID NO:1808;
(b) amino acids 100-127 of SEQ ID NO:1249; and (c) amino acids 98 to 127 of SEQ ID NO:1250;
wherein said fusion protein has GLP-1 activity.
9. The albumin fusion protein of claim 8, wherein said GLP-1 polypeptides comprise two amino acid sequences of (a).
10. The albumin fusion protein of claim 8, wherein said GLP-1 polypeptides comprise two amino acid sequences of (b).
11. The albumin fusion protein of claim 8, wherein said GLP-1 polypeptides comprise two amino acid sequences of (c).
12. The albumin fusion protein of claim 8, wherein said GLP-1 polypeptides comprise one amino acid sequence of (b) and one amino acid sequence of (c).
13. The albumin fusion protein of claim 8, wherein said GLP-1 polypeptides comprise one amino acid sequence of (a) and one amino acid sequence of (c).
14. An albumin fusion protein comprising two tandemly oriented GLP-1 polypeptides fused to albumin, wherein said albumin fusion protein comprises an amino acid sequence selected from:
(a) amino acids 25 to 669 of SEQ ID NO:1231;
(b) amino acids 25 to 669 of SEQ ID NO:1232;
(c) amino acids 25 to 669 of SEQ ID NO:1233;
(d) amino acids 25 to 667 of SEQ ID NO:1234;
(e) amino acids 25 to 669 of SEQ ID NO:1235;
(f) amino acids 25 to 669 of SEQ ID NO:1236;
(g) amino acids 25 to 667 of SEQ ID NO:1237;
(h) amino acids 30 to 674 of SEQ ID NO:1280;
(i) amino acids 20 to 664 of SEQ ID NO:1607;
(j) amino acids 20 to 664 of SEQ ID NO:1608;
(k) amino acids 19 to 663 of SEQ ID NO:1609;
(1) amino acids 19 to 663 of SEQ ID NO:1610;
(m) amino acids 24 to 668 of SEQ ID NO:1621;

(n) amino acids 86 to 730 of SEQ ID NO:1622;
(o) amino acids 18 to 662 of SEQ ID NO:1623;
(p) amino acids 86 to 730 of SEQ ID NO:1624;
(q) amino acids 24 to 668 of SEQ ID NO:1625;
(r) amino acids 18 to 662 of SEQ ID NO:1626; and (s) amino acids 30 to 673 of SEQ ID NO:2170;
and wherein said fusion protein has GLP-1 activity.
15. An albumin fusion protein comprising two tandemly oriented GLP-1 (7-36(A8G) polypeptides fused to albumin, wherein said fusion protein is produced from a host cell comprising a construct which expresses said albumin fusion protein, wherein said construct is pSAC35:KT.GLP-1(7-36(A8G))x2.HSA contained in ATCC Deposit No.
PTA-4671.
16 The albumin fusion protein of any one of claims 1 to 6, 8 to 12 and 13, wherein said GLP-1 polypeptides are fused at the N-terminus or at the C-terminus to albumin.
17. A polynucleotide encoding the albumin fusion protein of any one of claims 1 to 16.
18. The albumin fusion protein of any one of claims 1 to 16, which is glycosylated.
19. The albumin fusion protein of any one of claims 1 to 16, which is non-glycosylated.
20. The albumin fusion protein of any one of claims 1 to 16, which is expressed in yeast.
21. The albumin fusion protein of claim 20, wherein said yeast is a S.
cerevisiae, a K. lactis or a P. pastoris.
22. The albumin fusion protein of claim 20 or 21, wherein said yeast is glycosylation deficient.
23. The albumin fusion protein of any one of claims 20 to 22, wherein said yeast is glycosylation and protease deficient.
24. The albumin fusion protein of any one of claims 1 to 16, which is expressed by a mammalian cell.
25. The albumin fusion protein of claim 24, wherein said mammalian cell is a CHO cell, a COS cell or an NSO cell.
26. The albumin fusion protein of any one of claims 1 to 16, 18 to 24 and 25, wherein the albumin fusion protein further comprises a secretion leader sequence.
27. A composition comprising the albumin fusion protein of any one of claims 1 to 16 and 18 to 25, and a pharmaceutically acceptable carrier.
28. Use of the albumin fusion protein of claim 1, in the preparation of a pharmaceutical composition for treatment of hyperglycemia; diabetes; diabetes insipidus;
diabetes mellitus; type 1 diabetes; type 2 diabetes; insulin resistance;
insulin deficiency;
hyperlipidemia; hyperketonemia; non-insulin dependent diabetes mellitus (NIDDM);
insulin-dependent diabetes mellitus (IDDM); obesity; congestive heart failure;
or syndrome X.
29. Use of a therapeutically effective amount of the albumin fusion protein of any one of claims 1 to 16, 18 to 25 and 26, for treating hyperglycemia;
diabetes; diabetes insipidus; diabetes mellitus; type 1 diabetes; type 2 diabetes; insulin resistance; insulin deficiency; hyperlipidemia; hyperketonemia; non-insulin dependent diabetes mellitus (NIDDM); insulin-dependent diabetes mellitus (IDDM); obesity; congestive heart failure;
or syndrome X, in a patient.
30. The use according to claim 28 or 29, wherein said condition(s) associated with diabetes is obesity, heart disease, hyperglycemia, infection, retinopathy and/or ulcer.
31. Use of the albumin fusion protein of claim 1, in the preparation of a pharmaceutical composition for stimulation of insulin synthesis and release, to enhance adipose, muscle or liver tissue sensitivity towards insulin uptake, to stimulate glucose uptake, to slow digestive process, or to block the secretion of glucagon in a patient.
32. Use of a therapeutically effective amount of the albumin fusion protein of claim 1, to stimulate insulin synthesis and release, to enhance adipose, muscle or liver tissue sensitivity towards insulin uptake, stimulating glucose uptake, to slow digestive process, or to block the secretion of glucagon in a patient.
33. The use according to any one of claims 28 to 32, wherein said tandemly oriented GLP-1 polypeptides are selected from:
(i) one wild-type GLP-1 polypeptide fused to one GLP-1 fragment polypeptide;
(ii) one wild-type GLP-1 polypeptide fused to one GLP-1 variant polypeptide;
and (iii) one GLP-1 fragment polypeptide fused to one GLP-1 variant polypeptide.
34. The use according to any one of claims 28 to 32, wherein said GLP-1 fragments or GLP-1 variants are selected from:
(a) GLP-1 (9-36);
(b) GLP-1 (7-36); and (c) GLP-1 (7-36(A8G)).
35. The use according to claim 34, wherein said GLP-1 fragments or GLP-1 variants are selected from two tandemly oriented GLP-1 (7-36(A8G)).
36. The use according to any one of claims 28 to 32, wherein said albumin fusion protein is produced from a host cell comprising a construct which expresses said albumin fusion protein, wherein said construct is selected from the following constructs:
(a) pSAC:GLP-1(7-36)x2.FISA;
(b) pSAC35:GLP-1(7-36)x2.HSA;
(c) pSAC35:GLP-1(7-36(A8G))x2.HSA;
(d) pSAC35:coGLP-1(7-36(A8G))x2.HSA;
(e) pSAC35:GLP-1(7-36(A8S)).GLP-1(7-36).HSA;
(f) pSAC35:GLP-1(7-36(A8G).GLP-1(7-36).HSA;
(g) pSAC35:KT.GLP-1(7-36(A8G))x2.HSA;
(h) pSAC35:INV.GLP-1(7-36A8G)x2.HSA;
(i) pSAC35:INV.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(j) pSAC35:DeltaKex.GLP-1(7-36A8G)x2.HSA;
(k) pSAC35:DeltaKex.GLP-1(7-36A8G).GLP-1(7-36).HSA;
(l) pSAC35:INU.GLP-1(7-36(A8G))x2.HSA;
(m) pSAC35:MAF.GLP-1(7-36(A8G))x2.HSA;
(n) pSAC35:AP.GLP-1(7-36(A8G))x2.HSA;
(o) pSAC35:MAF.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(p) pSAC35:INU.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(q) pSAC35:AP.GLP-1(7-36(A8G)).GLP-1(7-36).HSA;
(r) pSAC:KT.GLP-1(7-36(A8G))x2.MSA.E25-A608; and (s) pSAC35:GLP-1(9-36).GLP-1(7-36).HSA.
37. The use according to any one of claims 28 to 32, wherein said albumin fusion protein comprises an amino acid sequence selected from:
(a) amino acids 1 to 30 of SEQ ID NO:1808;
(b) amino acids 100-127 of SEQ ID NO:1249; and (c) amino acids 98 to 127 of SEQ ID NO:1250;

wherein said fusion protein has GLP-1 activity.
38. The use according to claim 37, wherein said GLP-1 polypeptides comprise:
(i) two amino acid sequences of (a);
(ii) two amino acid sequences of (b);
(iii) two amino acid sequences of (c);
(iv) one amino acid sequence of (b) and one amino acid sequence of (c); or (v) one amino acid sequence of (a) and one amino acid sequence of (c).
39. The use according to any one of claims 28 to 32, wherein said albumin fusion protein comprises an amino acid sequence selected from:
(a) amino acids 25 to 669 of SEQ ID NO:1231;
(b) amino acids 25 to 669 of SEQ ID NO:1232;
(c) amino acids 25 to 669 of SEQ ID NO:1233;
(d) amino acids 25 to 667 of SEQ ID NO:1234;
(e) amino acids 25 to 669 of SEQ ID NO:1235;
(f) amino acids 25 to 669 of SEQ ID NO:1236;
(g) amino acids 25 to 667 of SEQ ID NO:1237;
(h) amino acids 30 to 674 of SEQ ID NO:1280;
(i) amino acids 20 to 664 of SEQ ID NO:1607;
(j) amino acids 20 to 664 of SEQ ID NO:1608;
(k) amino acids 19 to 663 of SEQ ID NO:1609;
(l) amino acids 19 to 663 of SEQ ID NO:1610;
(m) amino acids 24 to 668 of SEQ ID NO:1621;
(n) amino acids 86 to 730 of SEQ ID NO:1622;
(o) amino acids 18 to 662 of SEQ ID NO:1623;
(p) amino acids 86 to 730 of SEQ ID NO:1624;
(q) amino acids 24 to 668 of SEQ ID NO:1625;
(r) amino acids 18 to 662 of SEQ ID NO:1626; and (s) amino acids 30 to 673 of SEQ ID NO:2170;
and wherein said fusion protein has GLP-1 activity.
40. The use according to any one of claims 28 to 32, wherein said fusion protein comprises two tandemly oriented GLP-1 (7-36(A8G)) and is produced from a host cell comprising a construct which expresses said fusion protein, wherein said construct is pSAC35:KT.GLP-1(7-36(A8G))x2.HSA construct contained in ATCC Deposit No. PTA-4671.
41. The use according to any one of claims 28 to 35, 37 and 38, wherein said GLP-1 polypeptides are fused at the N-terminus or at the C-terminus to albumin.
42. The use according to any one of claims 28 to 41, wherein said albumin fusion protein is glycosylated.
43. The use according to any one of claims 28 to 41, wherein said albumin fusion protein is non-glycosylated.
44. The use according to any one of claims 28 to 41, wherein said albumin fusion protein is expressed in yeast.
45. The use according to claim 44, wherein said yeast is S. cerevisiae, K
lactis or P. pastoris.
46. The use according to claim 44 or 45, wherein said yeast is glycosylation deficient.
47. The use according to any one of claims 44 to 46, wherein said yeast is glycosylation and protease deficient.
48. The use according to any one of claims 28 to 41, wherein said albumin fusion protein is expressed by a mammalian cell.
49. The use according to claim 48, wherein said mammalian cell is a CHO
cell, a COS cell or an NSO cell.
50. The use according to any one of claims 28 to 49, wherein said albumin fusion protein further comprises a secretion leader sequence.
51. The use according to any one of claims 28 to 50, wherein the use further comprises use of a pharmaceutically acceptable carrier.
52. Use of the albumin fusion protein of claim 1 in the preparation of a pharmaceutical composition for suppression of body weight or suppression of appetite.
53. Use of the albumin fusion protein of claim 1 for suppression of body weight or suppression of appetite.
CA2471363A 2001-12-21 2002-12-23 Albumin fusion proteins Expired - Fee Related CA2471363C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2841097A CA2841097A1 (en) 2001-12-21 2002-12-23 Albumin and g-csf fusion proteins

Applications Claiming Priority (43)

Application Number Priority Date Filing Date Title
US34181101P 2001-12-21 2001-12-21
US60/341,811 2001-12-21
US35035802P 2002-01-24 2002-01-24
US60/350,358 2002-01-24
US35136002P 2002-01-28 2002-01-28
US60/351,360 2002-01-28
US35937002P 2002-02-26 2002-02-26
US60/359,370 2002-02-26
US36000002P 2002-02-28 2002-02-28
US60/360,000 2002-02-28
US36750002P 2002-03-27 2002-03-27
US60/367,500 2002-03-27
US37022702P 2002-04-08 2002-04-08
US60/370,227 2002-04-08
US37895002P 2002-05-10 2002-05-10
US60/378,950 2002-05-10
US38261702P 2002-05-24 2002-05-24
US60/382,617 2002-05-24
US38312302P 2002-05-28 2002-05-28
US60/383,123 2002-05-28
US38570802P 2002-06-05 2002-06-05
US60/385,708 2002-06-05
US39462502P 2002-07-10 2002-07-10
US60/394,625 2002-07-10
US39800802P 2002-07-24 2002-07-24
US60/398,008 2002-07-24
US40213102P 2002-08-09 2002-08-09
US60/402,131 2002-08-09
US40270802P 2002-08-13 2002-08-13
US60/402,708 2002-08-13
US41135502P 2002-09-18 2002-09-18
US41142602P 2002-09-18 2002-09-18
US60/411,355 2002-09-18
US60/411,426 2002-09-18
US41498402P 2002-10-02 2002-10-02
US60/414,984 2002-10-02
US41761102P 2002-10-11 2002-10-11
US60/417,611 2002-10-11
US42024602P 2002-10-23 2002-10-23
US60/420,246 2002-10-23
US42362302P 2002-11-05 2002-11-05
US60/423,623 2002-11-05
PCT/US2002/040891 WO2003060071A2 (en) 2001-12-21 2002-12-23 Albumin fusion proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CA2841097A Division CA2841097A1 (en) 2001-12-21 2002-12-23 Albumin and g-csf fusion proteins

Publications (2)

Publication Number Publication Date
CA2471363A1 CA2471363A1 (en) 2003-07-24
CA2471363C true CA2471363C (en) 2014-02-11

Family

ID=27586544

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2471363A Expired - Fee Related CA2471363C (en) 2001-12-21 2002-12-23 Albumin fusion proteins
CA2841097A Abandoned CA2841097A1 (en) 2001-12-21 2002-12-23 Albumin and g-csf fusion proteins

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA2841097A Abandoned CA2841097A1 (en) 2001-12-21 2002-12-23 Albumin and g-csf fusion proteins

Country Status (10)

Country Link
US (22) US7141547B2 (en)
EP (7) EP1997829A1 (en)
JP (9) JP5424521B2 (en)
AU (1) AU2002364586A1 (en)
CA (2) CA2471363C (en)
CY (2) CY1114265T1 (en)
DK (1) DK1463751T3 (en)
ES (3) ES2545090T3 (en)
PT (1) PT1463751E (en)
WO (1) WO2003060071A2 (en)

Families Citing this family (342)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US20060084082A1 (en) * 1997-03-07 2006-04-20 Human Genome Sciences, Inc. 186 human secreted proteins
US7393823B1 (en) 1999-01-20 2008-07-01 Oregon Health And Science University HER-2 binding antagonists
US7625859B1 (en) 2000-02-16 2009-12-01 Oregon Health & Science University HER-2 binding antagonists
US20030095967A1 (en) 1999-01-25 2003-05-22 Mackay Fabienne BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20050100991A1 (en) * 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
AU2001264563A1 (en) * 2000-04-12 2001-10-30 Human Genome Sciences, Inc. Albumin fusion proteins
US7632983B2 (en) 2000-07-31 2009-12-15 Biolex Therapeutics, Inc. Expression of monoclonal antibodies in duckweed
US8022270B2 (en) 2000-07-31 2011-09-20 Biolex Therapeutics, Inc. Expression of biologically active polypeptides in duckweed
DE60139430D1 (en) 2000-12-07 2009-09-10 Lilly Co Eli GLP-1 fusion proteins
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US7507413B2 (en) * 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
US20050123925A1 (en) 2002-11-15 2005-06-09 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20080194481A1 (en) 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
EP1997829A1 (en) * 2001-12-21 2008-12-03 Human Genome Sciences, Inc. Albumin fusion proteins
CA2484556A1 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
US20050222023A1 (en) * 2002-02-07 2005-10-06 Hans-Peter Hauser Albumin-fused kunitz domain peptides
US20060241027A1 (en) * 2002-02-07 2006-10-26 Hans-Peter Hauser Hiv inhibiting proteins
US20060122374A1 (en) * 2002-02-07 2006-06-08 Peter Mertins Albumin-fused anti-angiogenesis peptides
CN101172091B (en) * 2007-09-25 2011-04-27 北京美福源生物医药科技有限公司 Technique for preparing amalgamation protein skin-protection product containing albuminar and skin cell growth factor, and uses of the same
CN1241946C (en) * 2002-07-01 2006-02-15 美国福源集团 Human serum albumins recombined merge protein having hyperplasia stimulation function to multiple cells
US7731947B2 (en) 2003-11-17 2010-06-08 Intarcia Therapeutics, Inc. Composition and dosage form comprising an interferon particle formulation and suspending vehicle
CA2513213C (en) * 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
WO2004096113A2 (en) * 2003-04-28 2004-11-11 Medical Instill Technologies, Inc. Container with valve assembly for filling and dispensing substances, and apparatus and method for filling
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
US20070161087A1 (en) * 2003-05-29 2007-07-12 Wolfgang Glaesner Glp-1 fusion proteins
KR100758755B1 (en) * 2003-06-12 2007-09-14 일라이 릴리 앤드 캄파니 -1 glp-1 analog fusion proteins
US7176292B2 (en) * 2003-09-11 2007-02-13 Dialean, Ltd. Ghrelin variant protein
EP1680138B1 (en) * 2003-10-17 2013-07-17 Novo Nordisk A/S Combination therapy
CN1938333A (en) * 2003-12-03 2007-03-28 达尔塔生物技术有限公司 Interleukin-11 fusion proteins
PL1729795T3 (en) * 2004-02-09 2016-08-31 Human Genome Sciences Inc Albumin fusion proteins
EP1789440A4 (en) * 2004-02-11 2008-03-12 Amylin Pharmaceuticals Inc Pancreatic polypeptide family motifs and polypeptides comprising the same
WO2006097793A2 (en) 2004-04-15 2006-09-21 Chiasma, Ltd. Compositions capable of facilitating penetration across a biological barrier
US20070219131A1 (en) * 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
EP2348114B1 (en) 2004-04-21 2018-07-25 Alexion Pharmaceuticals, Inc. Bone delivery conjugates and method of using same to target proteins to bone
CA2564859C (en) * 2004-04-27 2013-04-09 Vital Therapies, Inc. Metabolic detoxification system and method
US8604185B2 (en) 2004-07-20 2013-12-10 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
EP2361931B1 (en) 2004-07-20 2017-12-06 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
RU2380411C2 (en) 2004-07-20 2010-01-27 Дженентек, Инк. Method for inhibition of hepatocyte proliferation, method for inhibition of hepatocyte cell adhesion and method for inhibition of biological activity angptl4 in hepatocytes or hepatocyte precursors
AU2005295713B2 (en) 2004-10-13 2011-06-16 The Washington University Use of BAFF to treat sepsis
US7572451B2 (en) 2004-10-25 2009-08-11 Cytos Biotechnology Ag Gastric inhibitory polypeptide (GIP) antigen arrays and uses thereof
GB0426146D0 (en) 2004-11-29 2004-12-29 Bioxell Spa Therapeutic peptides and method
EP1841796A2 (en) 2004-12-02 2007-10-10 Domantis Limited Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
WO2006067170A1 (en) * 2004-12-23 2006-06-29 Laboratoires Serono S.A. G-csf polypeptides and uses thereof
US11246913B2 (en) 2005-02-03 2022-02-15 Intarcia Therapeutics, Inc. Suspension formulation comprising an insulinotropic peptide
WO2006083761A2 (en) 2005-02-03 2006-08-10 Alza Corporation Solvent/polymer solutions as suspension vehicles
US7767799B2 (en) * 2005-06-29 2010-08-03 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Nucleic acid molecules encoding recombinant interferon-α2 (IFNα2) mutants
KR20080071119A (en) * 2005-08-12 2008-08-01 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
CN101282990B (en) * 2005-08-26 2013-04-03 阿雷斯贸易股份有限公司 Method for preparing glycosylated interferon-beta
WO2007049940A1 (en) * 2005-10-27 2007-05-03 Peptron Co., Ltd Bioactive substance-blood protein conjugate and stabilization of a bioactive substance using the same
HUE029798T2 (en) 2005-11-04 2017-03-28 Glaxosmithkline Llc Methods for administering hypoglycemic agents
BRPI0618449A2 (en) * 2005-11-10 2011-08-30 Receptor Biologix Inc methods for producing receptor and binder isoforms
US20080280328A1 (en) * 2005-11-18 2008-11-13 Novozymes A/S Glucoamylase Variants
JP2009517009A (en) * 2005-11-24 2009-04-30 ラボラトワ セローノ エス.エイ. Erythropoietin polypeptides and their use
AU2006323925B2 (en) * 2005-12-09 2012-08-02 Ares Trading S.A. Method for purifying FSH or a FSH mutant
US8841255B2 (en) * 2005-12-20 2014-09-23 Duke University Therapeutic agents comprising fusions of vasoactive intestinal peptide and elastic peptides
US20130172274A1 (en) 2005-12-20 2013-07-04 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
WO2007073486A2 (en) 2005-12-20 2007-06-28 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
MX2008008076A (en) * 2005-12-22 2008-11-28 Conjuchem Biotechnologies Inc Process for the production of preformed conjugates of albumin and a therapeutic agent.
US8759292B2 (en) 2006-02-03 2014-06-24 Prolor Biotech, Llc Long-acting coagulation factors and methods of producing same
US9249407B2 (en) 2006-02-03 2016-02-02 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US8304386B2 (en) * 2006-02-03 2012-11-06 Prolor Biotech, Inc. Long-acting growth hormone and methods of producing same
US20150038413A1 (en) 2006-02-03 2015-02-05 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US8946155B2 (en) 2006-02-03 2015-02-03 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US8450269B2 (en) 2006-02-03 2013-05-28 Prolor Biotech Ltd. Long-acting growth hormone and methods of producing same
WO2007092252A2 (en) * 2006-02-03 2007-08-16 Modigene Inc Long-acting polypeptides and methods of producing same
US8048848B2 (en) * 2006-02-03 2011-11-01 Prolor Biotech Ltd. Long-acting interferons and derivatives thereof and methods thereof
US9458444B2 (en) 2006-02-03 2016-10-04 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US8048849B2 (en) 2006-02-03 2011-11-01 Modigene, Inc. Long-acting polypeptides and methods of producing same
US20140113860A1 (en) 2006-02-03 2014-04-24 Prolor Biotech Ltd. Long-acting polypeptides and methods of producing and administering same
US10351615B2 (en) 2006-02-03 2019-07-16 Opko Biologics Ltd. Methods of treatment with long-acting growth hormone
US10221228B2 (en) 2006-02-03 2019-03-05 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US8476234B2 (en) * 2006-02-03 2013-07-02 Prolor Biotech Inc. Long-acting coagulation factors and methods of producing same
EP1816201A1 (en) 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
US20080261893A1 (en) * 2006-02-24 2008-10-23 Denise Barbut Topical corneal analgesia using neurotensin receptor agonists and synergistic neurotensin combinations without delaying wound healing
WO2007109354A2 (en) 2006-03-21 2007-09-27 Amylin Pharmaceuticals, Inc. Peptide-peptidase inhibitor conjugates and methods of using same
WO2007112940A2 (en) * 2006-03-31 2007-10-11 Ablynx N.V. Albumin-derived amino acid sequence, use thereof for increasing the half-life of therapeutic proteins and of other therapeutic compounds and entities, and constructs comprising the same
US9222728B2 (en) 2006-04-24 2015-12-29 Medinstill Development Llc Penetrable and resealable lyophilization device
MX2008014870A (en) 2006-05-30 2009-02-12 Intarcia Therapeutics Inc Two-piece, internal-channel osmotic delivery system flow modulator.
WO2007146038A2 (en) 2006-06-07 2007-12-21 Human Genome Sciences, Inc. Albumin fusion proteins
US9505823B2 (en) 2006-08-07 2016-11-29 TEV A Biopharmaceuticals USA, Inc. Albumin-insulin fusion proteins
ES2422864T3 (en) 2006-08-09 2013-09-16 Intarcia Therapeutics, Inc Osmotic release systems and piston units
JO2945B1 (en) * 2006-09-13 2016-03-15 سميث كلاين بيتشام كوربوريشن Methods For Administering Long-lasting Hypoglycemic Agents.
EP2423220B1 (en) 2007-01-30 2015-04-29 Epivax, Inc. Regulatory t cell epitopes, compositions and uses thereof
CN101663407B (en) * 2007-02-22 2017-08-08 健泰科生物技术公司 method for detecting inflammatory bowel disease
EP2132300B1 (en) * 2007-03-09 2011-01-12 Vectorlogics, Inc. Cells for adenovirus vector and protein production
AU2008225100A1 (en) * 2007-03-12 2008-09-18 Mayo Foundation For Medical Education And Research Topical corneal analgesia using neurotensin receptor agonists and synergistic neurotensin combinations without delaying wound healing
WO2008133908A2 (en) 2007-04-23 2008-11-06 Intarcia Therapeutics, Inc. Suspension formulations of insulinotropic peptides and uses thereof
US8114630B2 (en) * 2007-05-02 2012-02-14 Ambrx, Inc. Modified interferon beta polypeptides and their uses
EP2195034A2 (en) 2007-09-27 2010-06-16 Amylin Pharmaceuticals, Inc. Peptide-peptidase inhibitor conjugates and methods of making and using same
CA2702448A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying fc-fusion proteins
EP2203465A1 (en) * 2007-10-22 2010-07-07 Merck Serono S.A. Method for purifying an fc-containing protein
US20090186819A1 (en) * 2007-12-11 2009-07-23 Marieve Carrier Formulation of insulinotropic peptide conjugates
US20100297081A1 (en) * 2007-12-18 2010-11-25 Huang Yanshan Pharmaceutical formulation containing recombinant human serum albumin-interferon alpha fusion protein
EP2240155B1 (en) 2008-02-13 2012-06-06 Intarcia Therapeutics, Inc Devices, formulations, and methods for delivery of multiple beneficial agents
CN102046207B (en) 2008-03-31 2013-08-28 葛兰素集团有限公司 Drug fusions and conjugates
ES2525065T3 (en) * 2008-04-11 2014-12-17 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and their conjugates
US8476408B2 (en) * 2008-06-13 2013-07-02 Alize Pharma Sas Unacylated ghrelin and analogs as therapeutic agents for vascular remodeling in diabetic patients and treatment of cardiovascular disease
CN102131516B (en) 2008-06-27 2016-03-16 杜克大学 Comprise the therapeutic agent of elastin-like peptides
GB0815039D0 (en) * 2008-08-19 2008-09-24 Secr Defence Protein expression
KR101683314B1 (en) * 2008-09-17 2016-12-06 키아스마 인코포레이티드 Pharmaceutical compositions and related methods of delivery
EP2352526A1 (en) * 2008-09-19 2011-08-10 Protox Therapeutics Inc. Treating cancer stem cells using targeted cargo proteins
US8632986B2 (en) 2008-10-28 2014-01-21 Uladzimir A. Murauski Methods and kits for detection of toxemia
US8889618B2 (en) 2008-11-07 2014-11-18 The General Hospital Corporation C-terminal fragments of glucagon-like peptide-1 (GLP-1)
WO2010056732A1 (en) 2008-11-12 2010-05-20 Marv Enterprises Llc Utilization of stents for the treatment of blood borne carcinomas
CN102282168A (en) 2008-11-18 2011-12-14 梅里麦克制药股份有限公司 Human serum albumin linkers and conjugates thereof
PT2373681T (en) * 2008-12-10 2017-04-11 Glaxosmithkline Llc Pharmaceutical compositions of albiglutide
EA023344B1 (en) * 2009-01-16 2016-05-31 Тева Фармасьютикал Индастриз Лтд. Method for treating or preventing neutropenia or leukopenia or decreasing the incidence of infection as manifested by febrile neutropenia
AU2014202190B2 (en) * 2009-01-16 2016-03-17 Teva Pharmaceutical Industries Ltd. New Stable Formulations of Recombinant Human Albumin-Human Granulocyte Colony Stimulating Factor Fusion Protein
WO2010081494A1 (en) * 2009-01-19 2010-07-22 King Faisal Specialist Hospital And Research Centre A method of increasing protein expression in eukaryotic cells
WO2010087994A2 (en) 2009-01-30 2010-08-05 Whitehead Institute For Biomedical Research Methods for ligation and uses thereof
EP2396347B1 (en) 2009-02-11 2017-04-12 Albumedix A/S Albumin variants and conjugates
WO2010096394A2 (en) 2009-02-17 2010-08-26 Redwood Biosciences, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
WO2010107789A1 (en) * 2009-03-17 2010-09-23 Marv Enterprises Llc Sequential extracorporeal treatment of bodily fluids
UY32514A (en) 2009-03-27 2010-10-29 Glaxo Group Ltd FUSIONS AND CONJUGATES OF AN INSULINOTROPIC DRUG AND dAb WITH SEMIVIDA IMPROVED SERIES
EP2427486B1 (en) 2009-05-07 2015-02-25 Novozymes Biopharma DK A/S Method for purifying albumin
US9663778B2 (en) 2009-07-09 2017-05-30 OPKO Biologies Ltd. Long-acting coagulation factors and methods of producing same
PL2464370T3 (en) 2009-08-14 2017-08-31 Phasebio Pharmaceuticals, Inc. Modified vasoactive intestinal peptides
CN101993485B (en) 2009-08-20 2013-04-17 重庆富进生物医药有限公司 Peptide analog homologous dimer capable of accelerating insulin secretion and application thereof
EP3222287A1 (en) 2009-08-24 2017-09-27 Amunix Operating Inc. Coagulation factor ix compositions and methods of making and using same
US8451450B2 (en) * 2009-09-14 2013-05-28 Bio-Rad Laboratories, Inc. Near real time optical phase conjugation
WO2011037623A1 (en) 2009-09-28 2011-03-31 Intarcia Therapeutics, Inc. Rapid establishment and/or termination of substantial steady-state drug delivery
SG10201406063XA (en) 2009-09-30 2014-11-27 Glaxo Group Ltd Drug fusions and conjugates with extended half life
BR112012009450A2 (en) 2009-10-30 2017-05-23 Novozymes Biopharma Dk As albumin variants
EP2494050A4 (en) * 2009-10-30 2013-10-30 Merck Sharp & Dohme Granulocyte-colony stimulating factor produced in glycoengineered pichia pastoris
EP2506868B1 (en) 2009-12-06 2017-11-15 Bioverativ Therapeutics Inc. Factor viii-fc chimeric and hybrid polypeptides, and methods of use thereof
CN102711795A (en) 2009-12-08 2012-10-03 泰华制药工业有限公司 BChE albumin fusions for the treatment of cocaine abuse
KR20130070576A (en) * 2010-04-09 2013-06-27 노보자임스 바이오파마 디케이 에이/에스 Albumin derivatives and variants
EP2566502A4 (en) 2010-05-04 2013-10-09 Glaxosmithkline Llc Methods for treating or preventing cardiovascular disorders and providing cardiovascular protection
NZ605400A (en) 2010-07-09 2015-05-29 Biogen Idec Hemophilia Inc Chimeric clotting factors
UA115120C2 (en) 2010-07-09 2017-09-25 Байоджен Хемофіліа Інк. Factor ix polypeptides and methods fo use thereof
WO2012012271A2 (en) * 2010-07-19 2012-01-26 Sialix, Inc. Novel glycosylated polypeptides
BR112013007385B1 (en) 2010-09-28 2022-07-26 Amylin Pharmaceuticals, Llc CHIMERIC POLYPEPTIDE, ITS USE AND COMPOSITION THAT COMPRISES IT
US8892184B2 (en) 2010-10-18 2014-11-18 Siemens Medical Solutions Usa, Inc. Systems and methods for reducing interference in a dual modality imaging system
US20130225496A1 (en) 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants
CN102453095A (en) * 2010-11-01 2012-05-16 天津药物研究院 Fusion protein containing GLP (glucagon-like peptide)-1 and preparation method and application thereof
CA2823066A1 (en) 2010-12-27 2012-07-05 Alexion Pharma International Sarl Compositions comprising natriuretic peptides and methods of use thereof
CN103415621A (en) 2011-01-14 2013-11-27 雷德伍德生物科技股份有限公司 Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
BR112013020383A2 (en) * 2011-02-09 2017-06-13 Glaxosmithkline Llc lyophilized formulations.
US20120208755A1 (en) 2011-02-16 2012-08-16 Intarcia Therapeutics, Inc. Compositions, Devices and Methods of Use Thereof for the Treatment of Cancers
US9273116B2 (en) 2011-02-28 2016-03-01 Korea University Research And Business Foundation Fusion protein comprising albumin and retinol-binding protein
US10064915B2 (en) 2011-02-28 2018-09-04 Korea University Research And Business Foundation Fusion protein comprising albumin and retinol-binding protein
US9701732B2 (en) 2011-02-28 2017-07-11 Korea University Research And Business Foundation Fusion protein comprising albumin and retinol-binding protein
WO2012116453A1 (en) 2011-03-03 2012-09-07 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US9089312B2 (en) 2011-03-10 2015-07-28 Western New England University Tamponade for biopsy surgery and method of operation
US9205131B2 (en) * 2011-03-30 2015-12-08 The Regents Of The University Of California Use of IL-17D for the treatment and prevention of cancers
WO2012136790A1 (en) 2011-04-07 2012-10-11 Glaxo Group Limited Compositions comprising fusion proteins or conjugates with an improved half -life
WO2012136792A2 (en) 2011-04-07 2012-10-11 Glaxo Group Limited Cck compositions
US8558546B2 (en) * 2011-04-13 2013-10-15 Mark Griswold Relaxometry
US8822417B2 (en) 2011-05-05 2014-09-02 Novozymes Biopharma DIC A/S Albumin variants
WO2012158493A2 (en) 2011-05-19 2012-11-22 Geysen Hendrik M Compounds that bind to the erythropoietin receptor
CA2873553C (en) 2011-06-06 2020-01-28 Phasebio Pharmaceuticals, Inc. Use of modified vasoactive intestinal peptides in the treatment of hypertension
EP3527218A1 (en) 2011-06-10 2019-08-21 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof
WO2012174056A1 (en) * 2011-06-13 2012-12-20 Neumedicines, Inc. Mitigation of cutaneous injury with il-12
CN103764665A (en) 2011-06-28 2014-04-30 怀特黑德生物医学研究所 Using sortases to install click chemistry handles for protein ligation
EP2729492B1 (en) 2011-07-05 2019-01-02 Albumedix Ltd Albumin formulation and use
EP2729157B1 (en) 2011-07-06 2019-01-16 The General Hospital Corporation A pentapeptide derived from the c-terminus of glucagon-like peptide 1 (glp-1) for use in treatment
PT3513804T (en) 2011-07-08 2022-06-02 Bioverativ Therapeutics Inc Factor viii chimeric and hybrid polypeptides, and methods of use thereof
CN104271588B (en) * 2011-07-08 2017-10-10 安米林药品有限责任公司 The engineered polypeptide of immunogenicity with enhanced acting duration and reduction
WO2013016454A1 (en) 2011-07-25 2013-01-31 Biogen Idec Hemophilia Inc. Assays to monitor bleeding disorders
CN103998470A (en) 2011-08-17 2014-08-20 科罗拉多大学董事会法人团体 Transferrin-tumstatin fusion protein and methods for producing and using the same
US20140193514A1 (en) * 2011-08-19 2014-07-10 Marv Enterprises Method for the Treatment of Cancer
US9730913B2 (en) * 2011-10-13 2017-08-15 The Johns Hopkins University High affinity beta lactamase inhibitors
US20140315817A1 (en) 2011-11-18 2014-10-23 Eleven Biotherapeutics, Inc. Variant serum albumin with improved half-life and other properties
US20150011431A1 (en) 2012-01-09 2015-01-08 The Scripps Research Institute Humanized antibodies
JP6684490B2 (en) 2012-01-09 2020-04-22 ザ・スクリップス・リサーチ・インスティテュート Ultralong complementarity determining regions and uses thereof
LT2804623T (en) 2012-01-12 2019-12-10 Bioverativ Therapeutics Inc Chimeric factor viii polypeptides and uses thereof
DK2822577T3 (en) 2012-02-15 2019-04-01 Bioverativ Therapeutics Inc RECOMBINANT FACTOR VIII PROTEINS
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
ES2771208T3 (en) 2012-02-15 2020-07-06 Bioverativ Therapeutics Inc Factor VIII compositions and methods of preparation and use thereof
SI2814844T1 (en) 2012-02-15 2017-10-30 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (trem-1)
SI2814842T1 (en) 2012-02-15 2018-10-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
EP2817626B1 (en) 2012-02-20 2017-09-27 Uladzimir A. Murauski Methods for detection of active malignancy
JP6441682B2 (en) 2012-03-16 2018-12-19 アルブミディクス リミティド Albumin variant
WO2013148405A2 (en) * 2012-03-27 2013-10-03 Felder Mitchell S Treatment for atherosclerosis
WO2013148117A1 (en) 2012-03-27 2013-10-03 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
WO2013151649A1 (en) 2012-04-04 2013-10-10 Sialix Inc Glycan-interacting compounds
WO2013157002A1 (en) 2012-04-19 2013-10-24 Prolor Biotech Inc. Long-acting oxyntomodulin variants and methods of producing same
US20150342897A1 (en) 2012-05-16 2015-12-03 Glaxo Group Limited Polypeptide loaded poca nanoparticles for oral administration
DK2849567T3 (en) 2012-05-17 2022-06-20 Extend Biosciences Inc CARRIERS FOR IMPROVED MEDICINE ADMINISTRATION
EP2851429B1 (en) 2012-05-18 2019-07-24 Adda Biotech Inc. Protein and protein conjugate for diabetes treatment, and applications thereof
US10052366B2 (en) 2012-05-21 2018-08-21 Alexion Pharmaceuticsl, Inc. Compositions comprising alkaline phosphatase and/or natriuretic peptide and methods of use thereof
AU2013270682A1 (en) 2012-06-08 2014-12-11 Biogen Ma Inc. Procoagulant compounds
EP2863940A4 (en) 2012-06-08 2016-08-10 Biogen Ma Inc Chimeric clotting factors
US10023628B2 (en) 2012-07-06 2018-07-17 Bioverativ Therapeutics Inc. Cell line expressing single chain factor VIII polypeptides and uses thereof
EA201792485A3 (en) 2012-07-11 2019-03-29 Амуникс Оперэйтинг Инк. COMPLEX OF FACTOR VIII WITH XTEN AND PROTEIN FACTOR VILLEBRAND VON AND ITS APPLICATION (OPTIONS)
AU2013289881B2 (en) 2012-07-13 2018-01-18 Zymeworks Bc Inc. Multivalent heteromultimer scaffold design and constructs
CN102816244A (en) * 2012-08-23 2012-12-12 无锡和邦生物科技有限公司 Fusion protein of exendin-4 peptide and human serum albumin (HSA) and preparation method thereof
US9345766B2 (en) 2012-08-30 2016-05-24 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-ERBB3 agents
TWI810729B (en) 2012-09-25 2023-08-01 美商百歐維拉提夫治療公司 Methods of using fix polypeptides
UA116217C2 (en) 2012-10-09 2018-02-26 Санофі Exendin-4 derivatives as dual glp1/glucagon agonists
AU2013331000B2 (en) 2012-10-18 2018-04-19 Bioverativ Therapeutics Inc. Methods of using a fixed dose of a clotting factor
EP2914293A4 (en) 2012-10-30 2016-04-20 Biogen Ma Inc Methods of using fviii polypeptide
MX2015005363A (en) 2012-11-08 2015-11-06 Novozymes Biopharma Dk As Albumin variants.
BR112015011583B1 (en) 2012-11-20 2023-03-14 Opko Biologics Ltd METHODS FOR INCREASE THE HYDRODYNAMIC SIZE OR VOLUME OF HUMAN GROWTH HORMONE, METHOD FOR INCREASE THE APPARENT MOLECULAR WEIGHT OF A POLYPEPTIDE, AND METHOD FOR INCREASE THE HALF-LIFE OF A POLYPEPTIDE
EP2931298A4 (en) * 2012-12-12 2016-07-06 Teva Pharma Fusion of human growth hormone and albumin, formulation and uses thereof
WO2014096149A1 (en) 2012-12-21 2014-06-26 Sanofi Exendin-4 Derivatives
US10259863B2 (en) 2013-01-11 2019-04-16 The California Institute For Biomedical Research Bovine fusion antibodies
WO2014113359A1 (en) 2013-01-15 2014-07-24 Teva Pharmaceutical Industries Ltd. Formulations of albu-bche, preparation and uses thereof
US9161966B2 (en) 2013-01-30 2015-10-20 Ngm Biopharmaceuticals, Inc. GDF15 mutein polypeptides
JP6272907B2 (en) 2013-01-30 2018-01-31 エヌジーエム バイオファーマシューティカルズ インコーポレイテッド Compositions and methods of use in the treatment of metabolic disorders
LT3889173T (en) 2013-02-15 2023-10-10 Bioverativ Therapeutics Inc. Optimized factor viii gene
WO2014125082A1 (en) 2013-02-16 2014-08-21 Novozymes Biopharma Dk A/S Pharmacokinetic animal model
EP2964666B1 (en) * 2013-03-06 2020-11-18 GlaxoSmithKline LLC Host cells and methods of use
PL2968470T3 (en) * 2013-03-12 2021-05-31 The General Hospital Corporation Modified mullerian inhibiting substance (mis) proteins and uses thereof for the treatment of diseases
EP2968477B1 (en) 2013-03-15 2019-12-04 Bioverativ Therapeutics Inc. Factor viii polypeptide formulations
JP6387392B2 (en) 2013-03-15 2018-09-05 バイオベラティブ セラピューティクス インコーポレイテッド Factor IX polypeptide preparation
US20140271538A1 (en) 2013-03-15 2014-09-18 Teva Pharmaceutical Industries Ltd. Recombinant Human Albumin-Human Granulocyte Colony Stimulating Factor for the Prevention of Neutropenia in Pediatric Patients
US9849066B2 (en) 2013-04-24 2017-12-26 Corning Incorporated Delamination resistant pharmaceutical glass containers containing active pharmaceutical ingredients
EP2994483A4 (en) * 2013-05-06 2017-02-08 Cell Machines, Inc. Methods and compositions related to large scale production of proteins
CN111518199A (en) 2013-07-18 2020-08-11 图鲁斯生物科学有限责任公司 Humanized antibodies with ultralong complementarity determining regions
US20160311885A1 (en) 2013-08-14 2016-10-27 Biogen Ma Inc. Recombinant factor viii proteins
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
WO2015041718A1 (en) 2013-09-20 2015-03-26 The General Hospital Corporation Uses of modified mullerian inhibiting substance (mis) proteins for the treatment of neurodegenerative diseases
EP3048899B1 (en) 2013-09-25 2021-09-08 Bioverativ Therapeutics Inc. On-column viral inactivation methods
CN103554266B (en) * 2013-10-11 2015-05-27 哈尔滨医科大学 Tumstatin fusogenic peptide with anti-tumor effect, and preparation method and application thereof
US20150158926A1 (en) 2013-10-21 2015-06-11 Opko Biologics, Ltd. Long-acting polypeptides and methods of producing and administering same
CN103656618B (en) * 2013-10-25 2016-06-08 广东药学院 The treatment polypeptide nano fiber gel preparation of skin wound, preparation method and application
EP3065769A4 (en) 2013-11-08 2017-05-31 Biogen MA Inc. Procoagulant fusion compound
EP4332839A2 (en) 2013-12-06 2024-03-06 Bioverativ Therapeutics Inc. Population pharmacokinetics tools and uses thereof
EP3079712B1 (en) 2013-12-11 2022-02-02 The General Hospital Corporation Use of mullerian inhibiting substance (mis) proteins for contraception and ovarian reserve preservation
EP3080154B1 (en) 2013-12-13 2018-02-07 Sanofi Dual glp-1/gip receptor agonists
TW201609795A (en) 2013-12-13 2016-03-16 賽諾菲公司 EXENDIN-4 peptide analogues as dual GLP-1/GIP receptor agonists
WO2015086730A1 (en) 2013-12-13 2015-06-18 Sanofi Non-acylated exendin-4 peptide analogues
WO2015086733A1 (en) 2013-12-13 2015-06-18 Sanofi Dual glp-1/glucagon receptor agonists
IL246476B (en) 2014-01-10 2022-06-01 Biogen Ma Inc Factor viii chimeric proteins and uses thereof
SG10201808249VA (en) 2014-03-24 2018-10-30 Bioverativ Therapeutics Inc Lyophilized factor ix formulations
TW201625670A (en) 2014-04-07 2016-07-16 賽諾菲公司 Dual GLP-1/glucagon receptor agonists derived from EXENDIN-4
TW201625669A (en) 2014-04-07 2016-07-16 賽諾菲公司 Peptidic dual GLP-1/glucagon receptor agonists derived from Exendin-4
TW201625668A (en) 2014-04-07 2016-07-16 賽諾菲公司 Exendin-4 derivatives as peptidic dual GLP-1/glucagon receptor agonists
EP3137899A2 (en) 2014-05-02 2017-03-08 Cerenis Therapeutics Holding SA Hdl therapy markers
WO2015172046A1 (en) 2014-05-08 2015-11-12 Phasebio Pharmaceuticals, Inc. Methods and compositions for treating cystic fibrosis
US9932381B2 (en) 2014-06-18 2018-04-03 Sanofi Exendin-4 derivatives as selective glucagon receptor agonists
WO2016004113A1 (en) 2014-06-30 2016-01-07 Biogen Ma Inc. Optimized factor ix gene
WO2016007873A1 (en) 2014-07-11 2016-01-14 The Regents Of The University Of Michigan Compositions and methods for treating craniosynostosis
FI3172232T3 (en) 2014-07-17 2024-03-19 Novo Nordisk As Site directed mutagenesis of trem-1 antibodies for decreasing viscosity.
MX370115B (en) 2014-07-30 2019-12-02 Ngm Biopharmaceuticals Inc Compositions and methods of use for treating metabolic disorders.
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
CR20170110A (en) 2014-09-26 2017-05-08 Bayer Pharma AG STABILIZED DERIVATIVES OF ADRENOMEDULINE AND USE OF THE SAME
US9889085B1 (en) 2014-09-30 2018-02-13 Intarcia Therapeutics, Inc. Therapeutic methods for the treatment of diabetes and related conditions for patients with high baseline HbA1c
EP3201225B1 (en) 2014-10-03 2020-08-19 NGM Biopharmaceuticals, Inc. Angptl8 polypeptide and its use for treatment of conditions associated with elevated triglycerides
ES2753391T3 (en) 2014-10-14 2020-04-08 Halozyme Inc Adenosine deaminase 2 (ADA2) compositions, variants thereof and methods of use thereof
EP3220961B1 (en) 2014-10-22 2023-07-05 Extend Biosciences, Inc. Therapeutic vitamin d conjugates
US9789197B2 (en) 2014-10-22 2017-10-17 Extend Biosciences, Inc. RNAi vitamin D conjugates
WO2016065052A1 (en) 2014-10-22 2016-04-28 Extend Biosciences, Inc. Insulin vitamin d conjugates
MA40835A (en) 2014-10-23 2017-08-29 Biogen Ma Inc ANTI-GPIIB / IIIA ANTIBODIES AND THEIR USES
MX357994B (en) 2014-10-31 2018-08-01 Ngm Biopharmaceuticals Inc COMPOSITIONS and METHODS OF USE FOR TREATING METABOLIC DISORDERS.
MA40861A (en) 2014-10-31 2017-09-05 Biogen Ma Inc ANTI-GLYCOPROTEIN IIB / IIIA ANTIBODIES
EP3218005B1 (en) 2014-11-12 2023-01-04 Seagen Inc. Glycan-interacting compounds and methods of use
US9879087B2 (en) 2014-11-12 2018-01-30 Siamab Therapeutics, Inc. Glycan-interacting compounds and methods of use
BR112017011900A2 (en) 2014-12-05 2018-02-27 Alexion Pharma Inc treatment of recombinant alkaline phosphatase attacks
JP6868561B2 (en) 2015-01-28 2021-05-12 アレクシオン ファーマシューティカルズ, インコーポレイテッド How to treat subjects with alkaline phosphatase deficiency
EP3253401A4 (en) 2015-02-03 2018-11-21 Chiasma Inc. Method of treating diseases
WO2016130518A2 (en) 2015-02-09 2016-08-18 Phasebio Pharmaceuticals, Inc. Methods and compositions for treating muscle disease and disorders
CN113598842A (en) 2015-06-03 2021-11-05 因塔西亚制药公司 Implant placement and removal system
AR105319A1 (en) 2015-06-05 2017-09-27 Sanofi Sa PROPHARMS THAT INCLUDE A DUAL AGONIST GLU-1 / GLUCAGON CONJUGATE HIALURONIC ACID CONNECTOR
PL3310347T3 (en) 2015-06-19 2021-12-27 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
TW201706291A (en) 2015-07-10 2017-02-16 賽諾菲公司 New EXENDIN-4 derivatives as selective peptidic dual GLP-1/glucagon receptor agonists
KR20180029262A (en) 2015-08-03 2018-03-20 바이오버라티브 테라퓨틱스 인크. Factor IX fusion proteins and methods for their manufacture and use
AU2016308624B2 (en) 2015-08-17 2022-06-23 Alexion Pharmaceuticals, Inc. Manufacturing of alkaline phosphatases
US10633428B2 (en) 2015-08-20 2020-04-28 Albumedix Ltd Albumin variants and conjugates
AU2016319540B2 (en) * 2015-09-08 2021-08-26 Jcr Pharmaceuticals Co., Ltd. Novel human serum albumin mutant
WO2017049132A1 (en) * 2015-09-18 2017-03-23 DNARx Systems and methods for nucleic acid expression in vivo
EP3355904A4 (en) 2015-09-28 2019-06-12 Alexion Pharmaceuticals, Inc. Identifying effective dosage regimens for tissue non-specific alkaline phosphatase (tnsalp)-enzyme replacement therapy of hypophosphatasia
CN106589130B (en) * 2015-10-14 2019-11-01 阿思科力(苏州)生物科技有限公司 A kind of Slit2D2-HSA recombinant protein and its application in treatment pyemia
US10618952B2 (en) * 2015-10-30 2020-04-14 Medimmune Limited Prevention of N-terminal truncation in IgG light chains
EP3368062A4 (en) 2015-10-30 2019-07-03 Alexion Pharmaceuticals, Inc. Methods for treating craniosynostosis in a patient
MX2018005061A (en) 2015-11-12 2019-02-28 Siamab Therapeutics Inc Glycan-interacting compounds and methods of use.
AU2017214378B2 (en) 2016-02-01 2023-05-04 Bioverativ Therapeutics Inc. Optimized Factor VIII genes
EP3426286A4 (en) 2016-03-08 2019-12-04 Alexion Pharmaceuticals, Inc. Methods for treating hypophosphatasia in children
JP2017165713A (en) 2016-03-14 2017-09-21 Jcrファーマ株式会社 Serum albumin-20K growth hormone fusion protein
JP7021099B2 (en) 2016-03-31 2022-02-18 エヌジーエム バイオファーマシューティカルス,インコーポレーテッド Bound protein and how to use it
WO2017173395A1 (en) 2016-04-01 2017-10-05 Alexion Pharmaceuticals, Inc. Methods for treating hypophosphatasia in adolescents and adults
CN109152820A (en) 2016-04-01 2019-01-04 阿雷克森制药公司 It is powerless with alkaline phosphatase enzyme treatment muscle
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
KR102449167B1 (en) 2016-05-06 2022-09-28 파세비오 파마수티컬스 인코포레이티드 ELP fusion protein for controlled sustained release
EP3458084B1 (en) 2016-05-16 2020-04-01 Intarcia Therapeutics, Inc Glucagon-receptor selective polypeptides and methods of use thereof
USD840030S1 (en) 2016-06-02 2019-02-05 Intarcia Therapeutics, Inc. Implant placement guide
USD860451S1 (en) 2016-06-02 2019-09-17 Intarcia Therapeutics, Inc. Implant removal tool
US10988744B2 (en) 2016-06-06 2021-04-27 Alexion Pharmaceuticals, Inc. Method of producing alkaline phosphatase
WO2018026742A1 (en) * 2016-08-01 2018-02-08 Askgene Pharma Inc. Novel antibody-albumin-drug conjugates (aadc) and methods for using them
US11116821B2 (en) 2016-08-18 2021-09-14 Alexion Pharmaceuticals, Inc. Methods for treating tracheobronchomalacia
CN110337590A (en) 2016-11-04 2019-10-15 奥胡斯大学 The identification and treatment of tumour characterized by neonatal Fc receptor is overexpressed
EP3541847A4 (en) 2016-11-17 2020-07-08 Seattle Genetics, Inc. Glycan-interacting compounds and methods of use
WO2018102743A1 (en) 2016-12-02 2018-06-07 Bioverativ Therapeutics Inc. Methods of treating hemophilic arthropathy using chimeric clotting factors
EP3548063A1 (en) 2016-12-02 2019-10-09 Bioverativ Therapeutics Inc. Methods of inducing immune tolerance to clotting factors
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
EP3554532A2 (en) 2016-12-13 2019-10-23 Defensin Therapeutics ApS Methods for treating inflammatory conditions of the lungs
US11518793B2 (en) 2016-12-14 2022-12-06 The General Hospital Corporation Mullerian inhibiting substance (MIS) proteins for ovarian and uterine oncoprotection, and ovarian reserve and uterine preservation
MA47163A (en) 2016-12-16 2019-11-06 Biogen Ma Inc PROTEOLYTICALLY STABILIZED ACTIVATED GROWTH DIFFERENTIATION FACTOR 11
EP3565580B1 (en) 2017-01-03 2024-03-06 i2o Therapeutics, Inc. Continuous administration of exenatide and co-adminstration of acetaminophen, ethinylestradiol or levonorgestrel
MX2019009063A (en) 2017-01-31 2019-10-21 Bioverativ Therapeutics Inc Factor ix fusion proteins and methods of making and using same.
US20180251522A1 (en) * 2017-02-21 2018-09-06 The Regents Of The University Of Michigan Il-22bp compositions and method for the treatment of disease therewith
BR112019018043A2 (en) 2017-03-03 2020-04-07 Seattle Genetics Inc cancer treatment method, and, antibody-drug conjugate
CN116392609A (en) 2017-03-23 2023-07-07 Dnarx公司 Systems and methods for in vivo nucleic acid expression
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
AU2018243320A1 (en) 2017-03-31 2019-10-10 Alexion Pharmaceuticals, Inc. Methods for treating hypophosphatasia (HPP) in adults and adolescents
EP3628009A4 (en) * 2017-05-19 2021-03-24 The Regents of The University of Colorado, A Body Corporate Compositions and methods for improving cognition
TW201920255A (en) 2017-08-09 2019-06-01 美商生物化學醫療公司 Nucleic acid molecules and uses thereof
AU2018318313A1 (en) 2017-08-18 2020-03-19 Neutrolis Inc.. Engineered DNase enzymes and use in therapy
WO2019081643A1 (en) * 2017-10-27 2019-05-02 Euro-Diagnostica Ab Secreted reporter-peptides for optimizing cell-based assays for analysis on immuno-assay platforms
US10774139B2 (en) 2017-11-10 2020-09-15 Ngm Biopharmaceuticals, Inc. ANGPTL8-binding agents and methods of use thereof
BR112020015228A2 (en) 2018-02-01 2020-12-29 Bioverativ Therapeutics Inc. USE OF LENTIVIRAL VECTORS THAT EXPRESS FACTOR VIII
EP3774861A1 (en) 2018-03-28 2021-02-17 Bristol-Myers Squibb Company Interleukin-2/interleukin-2 receptor alpha fusion proteins and methods of use
EP3773684A1 (en) 2018-03-30 2021-02-17 Alexion Pharmaceuticals, Inc. Manufacturing of glycoproteins
SG11202009625WA (en) 2018-04-02 2020-10-29 Bristol Myers Squibb Co Anti-trem-1 antibodies and uses thereof
CA3100007A1 (en) 2018-05-14 2019-11-21 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides and methods of use thereof
BR112020023167A2 (en) 2018-05-14 2021-02-09 Werewolf Therapeutics, Inc. activatable cytokine polypeptides and methods of using these
KR20210020030A (en) 2018-05-18 2021-02-23 바이오버라티브 테라퓨틱스 인크. How to treat hemophilia A
BR112020026512A2 (en) 2018-07-03 2021-04-06 Bristol-Myers Squibb Company FGF-21 FORMULATIONS
AU2019308089A1 (en) 2018-07-19 2021-01-21 D&D Pharmatech Inc. Pharmaceutical composition comprising polypeptide
KR20200135618A (en) 2019-05-23 2020-12-03 ㈜ 디앤디파마텍 Pharmaceutical composition for preventing or treating nonalcoholic fatty liver disease comprising a polypeptide
MX2021001599A (en) 2018-08-09 2021-07-02 Bioverativ Therapeutics Inc Nucleic acid molecules and uses thereof for non-viral gene therapy.
WO2020056242A1 (en) * 2018-09-14 2020-03-19 Billion Bottle Project Ultraviolet (uv) dosimetry
US20200115430A1 (en) 2018-10-11 2020-04-16 Intarcia Therapeutics, Inc. Human amylin analog polypeptides and methods of use
CN109762069B (en) * 2018-11-05 2022-01-14 广州领晟医疗科技有限公司 Fusion protein, pharmaceutical composition and application thereof
WO2020099949A1 (en) * 2018-11-14 2020-05-22 Sudershan Biotech Pvt Ltd Recombinant vector, host cell and process for production of human serum albumin
EP3891289A2 (en) 2018-12-06 2021-10-13 Bioverativ Therapeutics Inc. Use of lentiviral vectors expressing factor ix
CN109627343A (en) * 2018-12-27 2019-04-16 北京美福源生物医药科技有限公司 Long-acting cytokine gene derivative fusion protein
CN113840625A (en) 2019-01-29 2021-12-24 夏尔-Nps医药品有限公司 Parathyroid hormone variants
MX2021013766A (en) 2019-05-14 2022-02-21 Werewolf Therapeutics Inc Separation moieties and methods and use thereof.
CN114174326A (en) 2019-06-18 2022-03-11 拜耳公司 Long-term stable adrenomedullin analogues and uses thereof
WO2021044360A1 (en) 2019-09-06 2021-03-11 Novartis Ag Therapeutic fusion proteins
TW202126284A (en) 2019-09-30 2021-07-16 美商百歐維拉提夫治療公司 Lentiviral vector formulations
CA3157859A1 (en) * 2019-10-15 2021-04-22 Modernatx, Inc. Mrnas encoding immune modulating polypeptides and uses thereof
WO2021075526A1 (en) 2019-10-17 2021-04-22 Jcrファーマ株式会社 Method for producing fusion protein of serum albumin and growth hormone
JP2021070700A (en) 2019-10-30 2021-05-06 Jcrファーマ株式会社 Aqueous pharmaceutical compositions comprising fusion protein between serum albumin and growth hormone
AU2020393434A1 (en) * 2019-11-27 2022-06-09 Revelations Biotech Pvt Ltd Nucleic acids, vectors, host cells and methods for production of fructosyltransferase from aspergillus japonicus
CN114761553A (en) * 2019-11-27 2022-07-15 启示录生物技术私人有限公司 Nucleic acids, vectors, host cells and methods for producing beta-fructofuranosidase from aspergillus niger
EP4069200A1 (en) 2019-12-04 2022-10-12 Albumedix Ltd Methods and compositions produced thereby
CN115605502A (en) * 2020-04-14 2023-01-13 希内丽思有限公司(Kr) Recombinant fusion protein for preventing or treating fibrotic diseases
WO2022032187A1 (en) 2020-08-07 2022-02-10 Bristol-Myers Squibb Company Fgf21 combined with ccr2/5 antagonists for the treatment of fibrosis
CN112724259B (en) * 2020-11-16 2022-12-20 天津林达生物科技有限公司 Fusion protein of human serum albumin and interleukin 2 and application thereof
WO2022115597A1 (en) 2020-11-25 2022-06-02 Bristol-Myers Squibb Company Methods of treating liver diseases
US11141457B1 (en) 2020-12-28 2021-10-12 Amryt Endo, Inc. Oral octreotide therapy and contraceptive methods
WO2022159414A1 (en) 2021-01-22 2022-07-28 University Of Rochester Erythropoietin for gastroinfestinal dysfunction
CN112915106A (en) * 2021-02-05 2021-06-08 张虎山 Preparation and application of tumor immune microenvironment regulator
CN114539426B (en) * 2022-03-02 2023-07-07 山东仙普爱瑞科技股份有限公司 Fusion protein containing interferon alpha, recombinant strain expressing fusion protein and preparation method of recombinant strain

Family Cites Families (643)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1172046A (en) * 1915-07-30 1916-02-15 Henry Psencik Plow.
US5625041A (en) * 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
DE2449885C3 (en) * 1974-10-21 1980-04-30 Biotest-Serum-Institut Gmbh, 6000 Frankfurt Process for the production of chemically modified, long-life hemoglobin preparations as well as the modified hemoglobin preparation produced by this process
US4002531A (en) 1976-01-22 1977-01-11 Pierce Chemical Company Modifying enzymes with polyethylene glycol and product produced thereby
US4363877B1 (en) 1977-09-23 1998-05-26 Univ California Recombinant dna transfer vectors
US4264731A (en) * 1977-05-27 1981-04-28 The Regents Of The University Of California DNA Joining method
US4440859A (en) * 1977-05-27 1984-04-03 The Regents Of The University Of California Method for producing recombinant bacterial plasmids containing the coding sequences of higher organisms
US4407948A (en) 1977-09-23 1983-10-04 The Regents Of The University Of California Purification of nucleotide sequences suitable for expression in bacteria
US4283489A (en) 1977-09-23 1981-08-11 The Regents Of The University Of California Purification of nucleotide sequences suitable for expression in bacteria
US4366246A (en) 1977-11-08 1982-12-28 Genentech, Inc. Method for microbial polypeptide expression
US4263428A (en) 1978-03-24 1981-04-21 The Regents Of The University Of California Bis-anthracycline nucleic acid function inhibitors and improved method for administering the same
US4447538A (en) * 1978-04-19 1984-05-08 Regents Of The University Of California Microorganism containing gene for human chorionic somatomammotropin
US4652525A (en) * 1978-04-19 1987-03-24 The Regents Of The University Of California Recombinant bacterial plasmids containing the coding sequences of insulin genes
US4246731A (en) * 1978-06-12 1981-01-27 Miro Carl F Window frame assembly
US4342832A (en) 1979-07-05 1982-08-03 Genentech, Inc. Method of constructing a replicable cloning vehicle having quasi-synthetic genes
AU538665B2 (en) 1979-10-30 1984-08-23 Juridical Foundation, Japanese Foundation For Cancer Research Human interferon dna
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
NO164037C (en) 1980-01-08 1990-08-22 Biogen Inc PROCEDURE FOR PREPARING A POLYPEPTIDE OF INTERFERON ALFA (IFNALFA) TYPE.
IE57069B1 (en) 1980-04-03 1992-04-22 Biogen Inc Dna sequences,recombinant dna molecules and processes for producing human fibroblast interferon
US4414510A (en) 1980-05-28 1983-11-08 General Electric Company Low cost sensing system and method employing anistropic magneto-resistive ferrite member
DE3023787A1 (en) 1980-06-25 1982-01-21 Studiengesellschaft Kohle mbH, 4330 Mülheim METHOD FOR INCREASING THE INCORPORATION AND EXPRESSION OF GENETIC MATERIAL IN THE CORE OF INTACT CELLS BY LIPOSOME
US6610830B1 (en) 1980-07-01 2003-08-26 Hoffman-La Roche Inc. Microbial production of mature human leukocyte interferons
FR2490675B1 (en) 1980-09-25 1985-11-15 Genentech Inc MICROBIAL PRODUCTION OF HUMAN FIBROPLASTER INTERFERON
DE3169595D1 (en) 1980-11-10 1985-05-02 Gersonde Klaus Method of preparing lipid vesicles by ultrasonic treatment, the use of this method and apparatus for its application
WO1982002715A1 (en) 1981-02-04 1982-08-19 Sugano Haruo Human interferon-beta gene
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
NZ199722A (en) 1981-02-25 1985-12-13 Genentech Inc Dna transfer vector for expression of exogenous polypeptide in yeast;transformed yeast strain
JPS57149228A (en) 1981-03-11 1982-09-14 Ajinomoto Co Inc Novel erythropoietin and its preparation
US4792602A (en) 1981-06-19 1988-12-20 Cornell Research Foundation, Inc. Adaptors, and synthesis and cloning of proinsulin genes
CA1204682A (en) 1981-06-19 1986-05-20 Saran A. Narang Adaptors, and synthesis and cloning of proinsulin genes
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
US4714681A (en) 1981-07-01 1987-12-22 The Board Of Reagents, The University Of Texas System Cancer Center Quadroma cells and trioma cells and methods for the production of same
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
DE3129906C3 (en) 1981-07-24 1996-12-19 Schering Ag Paramagnetic complex salts, their preparation and agents for use in NMR diagnostics
US4647447A (en) 1981-07-24 1987-03-03 Schering Aktiengesellschaft Diagnostic media
IL66614A (en) 1981-08-28 1985-09-29 Genentech Inc Method of constructing a dna sequence encoding a polypeptide,microbial production of human serum albumin,and pharmaceutical compositions comprising it
AU561343B2 (en) 1981-10-19 1987-05-07 Genentech Inc. Human immune interferon by recombinant dna
EP0079739A3 (en) 1981-11-12 1984-08-08 The Upjohn Company Albumin-based nucleotides, their replication and use, and plasmids for use therein
EP0091527A3 (en) 1981-12-14 1984-07-25 The President And Fellows Of Harvard College Dna sequences, recombinant dna molecules and processes for producing human serum albumin-like polypeptides
JPS58118008A (en) 1982-01-06 1983-07-13 Nec Corp Data processor
EP0098876A1 (en) 1982-01-19 1984-01-25 Cetus Corporation Multiclass hybrid interferons
DE3374837D1 (en) 1982-02-17 1988-01-21 Ciba Geigy Ag Lipids in the aqueous phase
US4450103A (en) * 1982-03-01 1984-05-22 Cetus Corporation Process for recovering human IFN-β from a transformed microorganism
US4775622A (en) 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
DE3377363D1 (en) 1982-03-31 1988-08-18 Ajinomoto Kk Gene coding for interleukin-2 polypeptide, recombinant dna carrying said gene, cell lines possessing the recombinant dna,and method for producing interleukin-2 using said cells
CA1341562C (en) 1982-03-31 2007-11-27 Tadatsugu Taniguchi Gene coded for interleukin-2 polypeptide, recombinant dna carrying the said gene, a living cell line possessing the recombinant dna, and method for producing interleukin-2 using the said cell
US4778879A (en) 1982-04-20 1988-10-18 Sloan-Kettering Institute For Cancer Research Highly purified human interleukin 2 and method
US4925919A (en) * 1984-04-25 1990-05-15 Roland Mertelsmann Purified interleukin 2
US5824330A (en) 1982-04-20 1998-10-20 Sloan-Kettering Institute For Cancer Research Highly purified interleukin-2 and method
US4499188A (en) * 1982-05-05 1985-02-12 Cetus Corporation Bacterial production of heterologous polypeptides under the control of a repressible promoter-operator
US6936694B1 (en) 1982-05-06 2005-08-30 Intermune, Inc. Manufacture and expression of large structural genes
DE3218121A1 (en) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Pharmaceutical compositions for tumour treatment
EP0102324A3 (en) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipids and surfactants in an aqueous medium
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
JPS5945835U (en) 1982-09-16 1984-03-27 株式会社東芝 Arc extinguishing device for circuit breakers and breakers
US4490289A (en) 1982-09-16 1984-12-25 Hoffmann-La Roche Inc. Homogeneous human interleukin 2
US4992271A (en) 1982-09-23 1991-02-12 Cetus Corporation Formulation for lipophilic IL-2 proteins
US4462940A (en) 1982-09-23 1984-07-31 Cetus Corporation Process for the recovery of human β-interferon-like polypeptides
FI82266C (en) 1982-10-19 1991-02-11 Cetus Corp Process for Preparation of IL-2 Mutein
US4741900A (en) 1982-11-16 1988-05-03 Cytogen Corporation Antibody-metal ion complexes
EP0116201B1 (en) 1983-01-12 1992-04-22 Chiron Corporation Secretory expression in eukaryotes
US4840934A (en) * 1983-01-25 1989-06-20 Eleanor Roosevelt Institute For Cancer Research, Inc. Therapeutic method using T cell growth factor
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US5015575A (en) * 1983-04-07 1991-05-14 Chiron Corporation Hybrid DNA synthesis of insulin
US4914026A (en) * 1983-04-07 1990-04-03 Chiron Corporation Alpha factor leader sequence directed secretion of insulin
DK172738B1 (en) 1983-04-07 1999-06-21 Chiron Corp Method for expressing mature insulin and DNA structure for use in the method
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1984004330A1 (en) 1983-04-22 1984-11-08 Amgen Secretion of exogenous polypeptides from yeast
NZ207926A (en) 1983-04-25 1988-04-29 Genentech Inc Use of yeast #a#-factor to assist in expression of proteins heterologus to yeast
US5010003A (en) 1983-04-25 1991-04-23 Genentech, Inc. Use of yeast homologous signals to secrete heterologous proteins
CA1196863A (en) * 1983-06-08 1985-11-19 Mattheus F.A. Goosen Slow release injectable insulin composition
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
JPS6087792A (en) 1983-09-23 1985-05-17 ジェネックス・コーポレイション Variant control region
DE3486459D1 (en) 1983-09-26 1997-12-11 Udo Dr Med Ehrenfeld Means and product for the diagnosis and therapy of tumors and for the treatment of weaknesses in cellular and humoral immune defense
DE3474511D1 (en) 1983-11-01 1988-11-17 Terumo Corp Pharmaceutical composition containing urokinase
JPS60115528A (en) 1983-11-28 1985-06-22 Takeda Chem Ind Ltd Human interleukin-2 protein, its production and pharmacological composition containing the same
GB8334102D0 (en) 1983-12-21 1984-02-01 Searle & Co Interferons with cysteine pattern
NZ210501A (en) * 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence
US4855238A (en) 1983-12-16 1989-08-08 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
GB8334261D0 (en) 1983-12-22 1984-02-01 Bass Plc Fermentation processes
IT1185503B (en) 1984-01-11 1987-11-12 Univ New York HUMAN Erythropietine ODNA CLONES
US4908433A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Uses of interleukin-2
US4908434A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Process for preparing purified interleukin-2
CA1213537A (en) 1984-05-01 1986-11-04 Canadian Patents And Development Limited - Societe Canadienne Des Brevets Et D'exploitation Limitee Polypeptide expression method
DK58285D0 (en) 1984-05-30 1985-02-08 Novo Industri As PEPTIDES AND MANUFACTURING AND USING THEREOF
EP0172619A1 (en) 1984-06-20 1986-02-26 Takeda Chemical Industries, Ltd. Novel transformant and use thereof
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
JPS61227526A (en) 1984-07-25 1986-10-09 Chugai Pharmaceut Co Ltd Novel csf and method of collecting same
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4687659A (en) 1984-11-13 1987-08-18 Salutar, Inc. Diamide-DTPA-paramagnetic contrast agents for MR imaging
US4959314A (en) 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
IL77081A (en) 1984-12-04 1999-10-28 Genetics Inst Dna sequence encoding human erythropoietin process for the preparation thereof and a pharmaceutical composition of human erythropoietin
US4946787A (en) 1985-01-07 1990-08-07 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
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
US5049386A (en) 1985-01-07 1991-09-17 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
US4677195A (en) 1985-01-11 1987-06-30 Genetics Institute, Inc. Method for the purification of erythropoietin and erythropoietin compositions
US4970300A (en) 1985-02-01 1990-11-13 New York University Modified factor VIII
GB8504099D0 (en) 1985-02-18 1985-03-20 Wellcome Found Physiologically active substances
DK129385A (en) 1985-03-22 1986-09-23 Novo Industri As PEPTIDES AND PREPARATION THEREOF
FR2579224B1 (en) * 1985-03-25 1987-05-22 Genetica PROCESS FOR THE MICROBIOLOGICAL PREPARATION OF HUMAN SERUM-ALBUMIN
US4751180A (en) * 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
DE3679343D1 (en) 1985-03-28 1991-06-27 Chiron Corp EXPRESSION BY USING FUSION GENES FOR PROTEIN PRODUCTION.
DE3668186D1 (en) 1985-04-01 1990-02-15 Celltech Ltd TRANSFORMED MYELOMA CELL LINE AND METHOD USING THE SAME FOR EXPRESSING A GENE ENCODING AN EUKARYONTIC POLYPEPTIDE.
GB8510219D0 (en) 1985-04-22 1985-05-30 Bass Plc Isolation of fermentation products
KR870000428A (en) 1985-06-17 1987-02-18 . Method of manufacturing human serum albumin
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
US4810643A (en) 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US5102872A (en) * 1985-09-20 1992-04-07 Cetus Corporation Controlled-release formulations of interleukin-2
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
JPH0618778B2 (en) 1985-10-04 1994-03-16 中外製薬株式会社 Leukopenia treatment
JPS6296086A (en) 1985-10-21 1987-05-02 Agency Of Ind Science & Technol Composite plasmid
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
CA1295563C (en) 1985-11-01 1992-02-11 Robert T. Garvin Production of active proteins containing cystine residues
US5576195A (en) 1985-11-01 1996-11-19 Xoma Corporation Vectors with pectate lyase signal sequence
US5641663A (en) * 1985-11-06 1997-06-24 Cangene Corporation Expression system for the secretion of bioactive human granulocyte macrophage colony stimulating factor (GM-CSF) and other heterologous proteins from steptomyces
CA1295566C (en) 1987-07-21 1992-02-11 Robert T. Garvin Characterization and structure of genes for protease a and protease b from streptomyces griseus
US5013824A (en) * 1985-11-19 1991-05-07 Schering Corporation Human interleukin-4 peptides and conjugates thereof
JPH0780930B2 (en) 1985-12-26 1995-08-30 三井石油化学工業株式会社 Method for polymerizing α-olefin
JPH0645551B2 (en) 1986-01-07 1994-06-15 塩野義製薬株式会社 Interleukin-2 composition
EP0231819B1 (en) 1986-01-22 1992-04-01 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical agent for the treatment of myelogenous leukemia
ES2039209T5 (en) 1986-01-22 1996-07-16 Chugai Pharmaceutical Co Ltd PHARMACEUTICAL COMPOUND THAT PROMOTES THE RESTORATION OF HEMOPOYETIC CAPACITY.
US4885363A (en) 1987-04-24 1989-12-05 E. R. Squibb & Sons, Inc. 1-substituted-1,4,7-triscarboxymethyl-1,4,7,10-tetraazacyclododecane and analogs
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
DE3772785D1 (en) 1986-01-23 1991-10-17 Squibb & Sons Inc 1-SUBSTITUTED-4,7,10-TRISCARBOXYMETHYL-1,4,7,10-TETRAAZACYCLODODECAN AND ANALOG.
DE3604868A1 (en) * 1986-02-15 1987-08-20 Behringwerke Ag INSULINE DERIVATIVES, METHOD FOR THEIR PRODUCTION AND THEIR USE
FR2594846B1 (en) * 1986-02-21 1989-10-20 Genetica PROCESS FOR THE PREPARATION OF MATURE HUMAN ALBUMIN SERUM
EP0237019A3 (en) 1986-03-14 1988-03-09 Toray Industries, Inc. Interferon conjugate and production thereof using recombinant gene
IT1203758B (en) 1986-03-27 1989-02-23 Univ Roma CLONING AND EXPRESSION VECTORS OF HETEROLOGICAL GENES IN YEASTS AND YEASES TRANSFORMED WITH SUCH CARRIERS
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
DK179286D0 (en) 1986-04-18 1986-04-18 Nordisk Gentofte INSULIN PREPARATION
US4765980A (en) 1986-04-28 1988-08-23 International Minerals & Chemical Corp. Stabilized porcine growth hormone
US4859609A (en) 1986-04-30 1989-08-22 Genentech, Inc. Novel receptors for efficient determination of ligands and their antagonists or agonists
US5028422A (en) 1986-05-27 1991-07-02 Schering Corporation Treatment of basal cell carcinoma intralesionally with recombinant human alpha interferon
US5985599A (en) 1986-05-29 1999-11-16 The Austin Research Institute FC receptor for immunoglobulin
GB8615701D0 (en) 1986-06-27 1986-08-06 Delta Biotechnology Ltd Stable gene integration vector
IT1196484B (en) 1986-07-11 1988-11-16 Sclavo Spa YEAST EXPRESSION AND SECRETION VECTOR, USEFUL FOR THE PREPARATION OF HETEROLOGICAL PROTEINS
GR871067B (en) 1986-07-18 1987-11-19 Chugai Pharmaceutical Co Ltd Process for producing stable pharmaceutical preparation containing granulocyte colony stimulating factor
US4857467A (en) 1986-07-23 1989-08-15 Phillips Petroleum Company Carbon and energy source markers for transformation of strains of the genes Pichia
US4801575A (en) * 1986-07-30 1989-01-31 The Regents Of The University Of California Chimeric peptides for neuropeptide delivery through the blood-brain barrier
US5002764A (en) * 1986-08-12 1991-03-26 Schering Corporation Treatment of actinic keratoses with alpha2 interferon
GB8620926D0 (en) 1986-08-29 1986-10-08 Delta Biotechnology Ltd Yeast promoter
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4929442A (en) * 1986-09-26 1990-05-29 Exovir, Inc. Compositions suitable for human topical application including a growth factor and/or related materials
US5508031A (en) * 1986-11-21 1996-04-16 Cetus Oncology Corporation Method for treating biological damage using a free-radial scavenger and interleukin-2
AU624789B2 (en) 1987-02-19 1992-06-25 Amgen, Inc. Purified platelet-derived growth factor and methods for purification thereof
US4835260A (en) * 1987-03-20 1989-05-30 Genetics Institute, Inc. Erythropoietin composition
GB8707398D0 (en) * 1987-03-27 1987-04-29 Coopers Animal Health Biologically active molecules
BR8806573A (en) 1987-04-09 1989-10-31 Delta Biotechnology Ltd LIGHT VECTOR
EP0682110A1 (en) 1987-04-22 1995-11-15 Chiron Corporation Recombinant DNA encoding PDGF A-chain polypeptides
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5489425A (en) 1987-06-24 1996-02-06 The Dow Chemical Company Functionalized polyamine chelants
US4994560A (en) 1987-06-24 1991-02-19 The Dow Chemical Company Functionalized polyamine chelants and radioactive rhodium complexes thereof for conjugation to antibodies
ES2086445T3 (en) 1987-07-16 1996-07-01 Nycomed Imaging As AMINO CARBOXYLIC ACIDS AND THEIR DERIVATIVES.
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
JPH02476A (en) 1987-07-28 1990-01-05 Gist Brocades Nv Kluyveromyces as host cell
JPH0622784Y2 (en) 1987-08-04 1994-06-15 東洋シヤッター株式会社 Height adjustment device for movable center pillar for over door
JP2627899B2 (en) 1987-08-19 1997-07-09 株式会社 ビタミン研究所 Production method of gene-encapsulated liposome
SE459586B (en) 1987-09-14 1989-07-17 Mta Szegedi Biolog Koezponti THE STRUCTURES CODING FOR AUTHENTIC HUMAN SERUM ALBUMIN AND PROCEDURES FOR ITS PREPARATION
PT88641B (en) 1987-10-02 1993-04-30 Genentech Inc METHOD FOR PREPARING A VARIETY OF ADHESION
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
GB8725529D0 (en) * 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
JPH0811074B2 (en) 1987-10-30 1996-02-07 財団法人化学及血清療法研究所 Recombinant plasmid incorporating a gene encoding prealbumin and method for producing prealbumin using the same
GB8726953D0 (en) 1987-11-18 1987-12-23 Delta Biotechnology Ltd Yeast expression system
JP2791418B2 (en) 1987-12-02 1998-08-27 株式会社ミドリ十字 Method for producing heterologous protein, recombinant DNA, transformant
PT89484B (en) 1988-01-22 1994-03-31 Gen Hospital Corp GENE CLONED CODES OF FUSAO PROTEINS IG-CD4 AND ITS UTILIZATION
JPH01240191A (en) 1988-02-16 1989-09-25 Green Cross Corp:The Novel signal peptide functioning by yeast and secretory manifestation of foreign protein using same
JPH01215289A (en) 1988-02-22 1989-08-29 Toa Nenryo Kogyo Kk Production of normal human serum albumin a through gene recombination
JPH01302543A (en) 1988-02-26 1989-12-06 Fujitsu Ltd Tracking error detecting method and diffraction grating and optical detector used for said detecting method
US4999339A (en) * 1988-03-28 1991-03-12 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US5066489A (en) 1988-03-28 1991-11-19 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US5981485A (en) 1997-07-14 1999-11-09 Genentech, Inc. Human growth hormone aqueous formulation
US5096885A (en) * 1988-04-15 1992-03-17 Genentech, Inc. Human growth hormone formulation
US5096707A (en) * 1988-04-15 1992-03-17 The United States Of America As Represented By The Department Of Health And Human Services Flavone-8-acetic acid and interleukin-2 in a method of treating certain cancers
US5763394A (en) * 1988-04-15 1998-06-09 Genentech, Inc. Human growth hormone aqueous formulation
US5061488A (en) 1988-04-15 1991-10-29 The United States Of America As Represented Department Of Health & Human Services Flavone-8-acetic acid and interleukin-2 for cancer therapy
GB8809129D0 (en) 1988-04-18 1988-05-18 Celltech Ltd Recombinant dna methods vectors and host cells
IL89992A0 (en) 1988-04-25 1989-12-15 Phillips Petroleum Co Expression of human serum albumin in methylotrophic yeasts
IL89989A0 (en) 1988-04-25 1989-12-15 Phillips Petroleum Co Expression of human interleukin-2 in methylotrophic yeasts
US5126129A (en) * 1988-05-23 1992-06-30 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Cancer therapy using interleukin-2 and flavone compounds
DE58906966D1 (en) 1988-06-23 1994-03-24 Hoechst Ag Mini-proinsulin, its production and use.
ZA894792B (en) 1988-06-24 1991-04-24 Dow Chemical Co Macrocyclic bifunctional chelants,complexes thereof and their antibody conjugates
US5756065A (en) 1988-06-24 1998-05-26 The Dow Chemical Company Macrocyclic tetraazacyclododecane conjugates and their use as diagnostic and therapeutic agents
NZ229700A (en) 1988-06-24 1993-01-27 Dow Chemical Co Tetraazacyclododecane derivatives containing a linker/spacer moiety capable of forming antibody conjugates; complexes with radionuclides and conjugates of such compounds and complexes with antibodies or antibody fragments
US5274119A (en) 1988-07-01 1993-12-28 The Dow Chemical Company Vicinal diols
ATE135045T1 (en) * 1988-07-23 1996-03-15 Delta Biotechnology Ltd SECRETORY LEADER SEQUENCES
US4925648A (en) 1988-07-29 1990-05-15 Immunomedics, Inc. Detection and treatment of infectious and inflammatory lesions
FR2635115B1 (en) 1988-08-05 1992-04-03 Rhone Poulenc Sante PROCESS FOR THE PREPARATION OF HUMAN ALBUMIN SERUM FROM YEAST
FR2649991B2 (en) 1988-08-05 1994-03-04 Rhone Poulenc Sante USE OF STABLE DERIVATIVES OF PLASMID PKD1 FOR THE EXPRESSION AND SECRETION OF HETEROLOGOUS PROTEINS IN YEASTS OF THE GENUS KLUYVEROMYCES
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
WO1990001540A1 (en) 1988-08-11 1990-02-22 California Biotechnology Inc. Method for stabilizing heterologous protein expression and vectors for use therein
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
EP1892296A1 (en) 1988-09-02 2008-02-27 Dyax Corporation Generation and selection of recombinant varied binding proteins
US5260202A (en) 1988-09-07 1993-11-09 Delta Biotechnology Limited Fermentation method
JPH02227079A (en) 1988-10-06 1990-09-10 Tonen Corp Human serum albumin fragment
US5298243A (en) 1988-10-20 1994-03-29 Denki Kagaku Kogyo Kabushiki Kaisha Colony stimulating factor-gelatin conjugate
US5349052A (en) 1988-10-20 1994-09-20 Royal Free Hospital School Of Medicine Process for fractionating polyethylene glycol (PEG)-protein adducts and an adduct for PEG and granulocyte-macrophage colony stimulating factor
US5759802A (en) 1988-10-26 1998-06-02 Tonen Corporation Production of human serum alubumin A
JPH02117384A (en) 1988-10-26 1990-05-01 Tonen Corp Production of human serum albumin a by yeast host
DE68929151T2 (en) 1988-10-28 2000-07-20 Genentech Inc METHOD FOR DETECTING ACTIVE DOMAINS AND AMINO ACID RESIDUES IN POLYPEPTIDES AND HORMONE VARIANTS
US5342604A (en) 1988-10-31 1994-08-30 The Dow Chemical Company Complexes possessing ortho ligating functionality
US5696239A (en) 1988-10-31 1997-12-09 The Dow Chemical Company Conjugates possessing ortho ligating functionality and complexes thereof
US5256410A (en) 1988-12-01 1993-10-26 Schering Corporation Treatment of squamous cell carcinoma intralesionally with recombinant human alpha interferon
JP2989002B2 (en) 1988-12-22 1999-12-13 キリン―アムジエン・インコーポレーテツド Chemically modified granulocyte colony stimulating factor
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DK105489D0 (en) 1989-03-03 1989-03-03 Novo Nordisk As POLYPEPTIDE
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5705363A (en) * 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
DE68929217T2 (en) 1989-03-20 2000-11-30 Gen Hospital Corp INSULINOTROPES HORMON
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5693622A (en) 1989-03-21 1997-12-02 Vical Incorporated Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
AU5344190A (en) 1989-03-21 1990-10-22 Vical, Inc. Expression of exogenous polynucleotide sequences in a vertebrate
US5231020A (en) 1989-03-30 1993-07-27 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5328470A (en) 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
ATE79763T1 (en) 1989-04-11 1992-09-15 Boehringer Ingelheim Int USE OF AT LEAST ONE CYTOKINE IN THE MANUFACTURE OF A MEDICATION FOR THE SYSTEMIC TREATMENT OF PRAENEOPLASTIC LESIONS.
EP0395918A3 (en) 1989-04-13 1991-10-23 Vascular Laboratory, Inc. Plasminogen activator complex of pure pro-urokinase covalently bound by a disulfide bridge to human serum albumin
US5324844A (en) 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
GB8909916D0 (en) 1989-04-29 1989-06-14 Delta Biotechnology Ltd Polypeptides
ATE92107T1 (en) 1989-04-29 1993-08-15 Delta Biotechnology Ltd N-TERMINAL FRAGMENTS OF HUMAN SERUM ALBUMIN-CONTAINING FUSION PROTEINS.
US5766883A (en) * 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
CA2017379C (en) 1989-05-24 2002-06-25 Kenneth A. Thomas, Jr. Purification and characterization of a glioma-derived growth factor
US5808003A (en) 1989-05-26 1998-09-15 Perimmune Holdings, Inc. Polyaminocarboxylate chelators
ATE125869T1 (en) 1989-06-09 1995-08-15 Gropep Pty Ltd GROWTH HORMONE FUSION PROTEINS.
EP0479909B1 (en) 1989-06-29 1996-10-30 Medarex, Inc. Bispecific reagents for aids therapy
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
DE3924746A1 (en) * 1989-07-26 1991-01-31 Behringwerke Ag ERTHROPOIETIN (EPO) PEPTIDES AND ANTIBODIES THEREFOR
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
CU22222A1 (en) 1989-08-03 1995-01-31 Cigb PROCEDURE FOR THE EXPRESSION OF HETEROLOGICAL PROTEINS PRODUCED IN A FUSION FORM IN ESCHERICHIA COLI, ITS USE, EXPRESSION VECTORS AND RECOMBINANT STRAINS
EP0489116B1 (en) 1989-08-22 1994-04-06 Immunex Corporation Fusion proteins comprising gm-csf and il-3
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
US5436146A (en) 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
US6063373A (en) * 1989-09-19 2000-05-16 Maxim Pharmaceuticals, Inc. Enhanced activation of NK cells using an NK cell activator and a hydrogen peroxide scavenger or inhibitor
CA1340994C (en) 1989-09-21 2000-05-16 Rudolf Edgar Dr. Falk Treatment of conditions and disease
WO1991005052A1 (en) 1989-09-28 1991-04-18 Leningradsky Gosudarstvenny Universitet Method for obtaining a polypeptide with human-interleukin-2 activity, secreted by yeast cells, saccharomyces cerevisiae
FR2653020B1 (en) 1989-10-17 1993-03-26 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF LEUKEMIA.
US5667986A (en) * 1989-10-18 1997-09-16 Delta Biotechnology Limited Yeast promoter for expressing heterologous polypeptides
GB8924021D0 (en) 1989-10-25 1989-12-13 Celltech Ltd Recombinant dna method and vectors for the use therein
AU625013B2 (en) 1989-11-03 1992-06-25 Vanderbilt University Method of in vivo delivery of functioning foreign genes
US5676954A (en) 1989-11-03 1997-10-14 Vanderbilt University Method of in vivo delivery of functioning foreign genes
DK608589D0 (en) 1989-12-01 1989-12-01 Holm Arne CHEMICAL PROCEDURE
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5116944A (en) * 1989-12-29 1992-05-26 Neorx Corporation Conjugates having improved characteristics for in vivo administration
JPH03201987A (en) 1989-12-29 1991-09-03 Tonen Corp Human serum albumin fragment
US5780225A (en) 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
WO1991010737A1 (en) 1990-01-11 1991-07-25 Molecular Affinities Corporation Production of antibodies using gene libraries
DE69133566T2 (en) 1990-01-12 2007-12-06 Amgen Fremont Inc. Formation of xenogenic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
CA2073856C (en) 1990-01-24 2002-12-03 Douglas I. Buckley Glp-1 analogs useful for diabetes treatment
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
JPH04211375A (en) 1990-02-05 1992-08-03 Ajinomoto Co Inc Synthetic gene and production of human serum albumin using the synthetic gene
US5208018A (en) * 1990-03-19 1993-05-04 Brigham And Women's Hospital Treatment of cachexia with interleukin 2
FR2660863B1 (en) * 1990-04-17 1994-01-21 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR THE PREPARATION OF A PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF PRIMITIVE CANCERS OF THE POWDER.
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
AU7906691A (en) 1990-05-23 1991-12-10 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Adeno-associated virus (aav)-based eucaryotic vectors
GB9013017D0 (en) * 1990-06-11 1990-08-01 Mars Uk Ltd Compounds
AU650893B2 (en) 1990-07-10 1994-07-07 Boehringer Ingelheim International Gmbh O-glycosylated alpha-2 interferon
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5225341A (en) 1990-07-19 1993-07-06 The Regents Of The University Of California Biologically safe plant transformation system using a ds transposon
US5202239A (en) * 1990-08-07 1993-04-13 Scios Nova Inc. Expression of recombinant polypeptides with improved purification
US5071872A (en) 1990-08-14 1991-12-10 The Ohio State University Research Foundation Method for improving interleukin-2 activity using aci-reductone compounds
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
JP2938569B2 (en) 1990-08-29 1999-08-23 ジェンファーム インターナショナル,インコーポレイティド Method for producing xenogeneic immunoglobulin and transgenic mouse
IT1242149B (en) 1990-09-27 1994-02-16 Consiglio Nazionale Ricerche CODING NUCLEOTID SEQUENCE FOR A HUMAN PROTEIN WITH REGULATORY PROPERTIES OF ANGIOGENESIS
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
ATE175118T1 (en) 1990-10-05 1999-01-15 Medarex Inc TARGETED IMMUNOSTIMULATION WITH BISPECIFIC SUBSTANCES
JPH07108232B2 (en) 1990-10-09 1995-11-22 エム・ディ・リサーチ株式会社 Method for producing peptide or protein
JPH0638771Y2 (en) 1990-10-26 1994-10-12 株式会社カンセイ Airbag control device
EP0557300B1 (en) 1990-10-29 1997-11-19 Chiron Corporation Bispecific antibodies, method of production, and uses thereof
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
FR2668368B1 (en) 1990-10-30 1995-03-10 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR PREPARING A PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF MALIGNANT EPITHELIAL TUMORS.
US5353535A (en) 1990-11-05 1994-10-11 Plumly George W Floor type advertising apparatus
US5367080A (en) 1990-11-08 1994-11-22 Sterling Winthrop Inc. Complexing agents and targeting radioactive immunoreagents useful in therapeutic and diagnostic imaging compositions and methods
US5830452A (en) 1990-11-20 1998-11-03 Chiron Corporation Method for enhancing the anti-tumor therapeutic index of interleukin-2
WO1992009690A2 (en) 1990-12-03 1992-06-11 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
US5272070A (en) 1991-03-08 1993-12-21 Board Of Regents, The University Of Texas System Method for the preparation of cell lines producing Man3 GlcNac 2 asparagine-linked gylcans and cell lines produced thereby
EP0503583A1 (en) 1991-03-12 1992-09-16 Takeda Chemical Industries, Ltd. Composition for sustained release of erythropoietin
CA2064331C (en) 1991-03-28 2003-02-18 Marvin L. Bayne Vascular endothelial cell growth factor c subunit
JP3672306B2 (en) 1991-04-10 2005-07-20 ザ スクリップス リサーチ インスティテュート Heterodimeric receptor library using phagemids
EP0509841A3 (en) 1991-04-18 1993-08-18 Tonen Corporation Co-expression system of protein disulfide isomerase gene and useful polypeptide gene and process for producing the polypeptide using its system
CA2058820C (en) 1991-04-25 2003-07-15 Kotikanyad Sreekrishna Expression cassettes and vectors for the secretion of human serum albumin in pichia pastoris cells
EP0511011B1 (en) 1991-04-26 1996-10-23 Surface Active Limited Novel antibodies and methods for their use
US5330901A (en) 1991-04-26 1994-07-19 Research Corporation Technologies, Inc. Expression of human serum albumin in Pichia pastoris
US5646012A (en) 1991-04-30 1997-07-08 Rhone-Poulenc Rorer S.A. Yeast promoter and use thereof
FR2676070B1 (en) 1991-04-30 1994-09-30 Rhone Poulenc Rorer Sa YEAST PROMOTER AND ITS USE.
EP0519596B1 (en) 1991-05-17 2005-02-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
US5304473A (en) * 1991-06-11 1994-04-19 Eli Lilly And Company A-C-B proinsulin, method of manufacturing and using same, and intermediates in insulin production
CA2110799A1 (en) 1991-06-14 1992-12-23 Arnold H. Horwitz Microbially-produced antibody fragments and their conjugates
FR2677996B1 (en) 1991-06-21 1993-08-27 Rhone Poulenc Rorer Sa CLONING AND / OR EXPRESSION VECTORS PREPARATION AND USE.
US5844095A (en) 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
US5851795A (en) 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
WO1993000109A1 (en) 1991-06-28 1993-01-07 Genentech, Inc. Method of stimulating immune response using growth hormone
US5633146A (en) * 1991-07-02 1997-05-27 Rhone-Poulenc Rorer S.A. Method for producing recombinant proteins and host cells used therein
US5223408A (en) * 1991-07-11 1993-06-29 Genentech, Inc. Method for making variant secreted proteins with altered properties
JPH05292972A (en) 1991-07-29 1993-11-09 Tonen Corp Improved yeast expression system
CA2114112A1 (en) 1991-07-31 1993-02-18 David R. Hurwitz Transgenic protein production
US5723719A (en) 1991-08-08 1998-03-03 Health Research Inc. Transgenic mouse as model for diseases involving dopaminergic dysfunction
DE4126968A1 (en) 1991-08-14 1993-02-18 Detlev Prof Dr Med Ganten Transgenic rats that contain at least one human gene in their genome that is involved in blood pressure regulation
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5641670A (en) 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
US6270989B1 (en) 1991-11-05 2001-08-07 Transkaryotic Therapies, Inc. Protein production and delivery
NZ245015A (en) * 1991-11-05 1995-12-21 Transkaryotic Therapies Inc Delivery of human growth hormone through the administration of transfected cell lines encoding human growth hormone, which are physically protected from host immune response; the transfected cells and their production
AU665599B2 (en) 1991-11-08 1996-01-11 Hemosol Inc. Hemoglobins as drug delivery agents
DK1024191T3 (en) 1991-12-02 2008-12-08 Medical Res Council Preparation of autoantibodies displayed on phage surfaces from antibody segment libraries
US6348327B1 (en) * 1991-12-06 2002-02-19 Genentech, Inc. Non-endocrine animal host cells capable of expressing variant proinsulin and processing the same to form active, mature insulin and methods of culturing such cells
US5428139A (en) 1991-12-10 1995-06-27 The Dow Chemical Company Bicyclopolyazamacrocyclophosphonic acid complexes for use as radiopharmaceuticals
AU671093B2 (en) 1992-01-07 1996-08-15 Elan Pharmaceuticals, Inc. Transgenic animal models for alzheimer's disease
GB9200417D0 (en) 1992-01-09 1992-02-26 Bagshawe Kenneth D Cytotoxic drug therapy
FR2686620B1 (en) * 1992-01-27 1995-06-23 Rhone Poulenc Rorer Sa HUMAN SERUM-ALBUMIN, PREPARATION AND USE.
FR2686901A1 (en) 1992-01-31 1993-08-06 Rhone Poulenc Rorer Sa NOVEL ANTITHROMBOTIC POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2686899B1 (en) * 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2686900B1 (en) * 1992-01-31 1995-07-21 Rhone Poulenc Rorer Sa NOVEL POLYPEPTIDES HAVING GRANULOCYTE COLONY STIMULATION ACTIVITY, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
ATE239506T1 (en) 1992-03-05 2003-05-15 Univ Texas USE OF IMMUNOCONJUGATES FOR THE DIAGNOSIS AND/OR THERAPY OF VASCULARIZED TUMORS
US5230886A (en) 1992-03-18 1993-07-27 Trustees Of Boston University Tumor cell suppression
USRE37302E1 (en) 1992-03-19 2001-07-31 Novo Nordisk A/S Peptide
DK36392D0 (en) * 1992-03-19 1992-03-19 Novo Nordisk As USE OF CHEMICAL COMPOUND
DK36492D0 (en) 1992-03-19 1992-03-19 Novo Nordisk As PREPARATION
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5460954A (en) 1992-04-01 1995-10-24 Cheil Foods & Chemicals, Inc. Production of human proinsulin using a novel vector system
US5229109A (en) 1992-04-14 1993-07-20 Board Of Regents, The University Of Texas System Low toxicity interleukin-2 analogues for use in immunotherapy
US5775745A (en) 1992-06-09 1998-07-07 Hoppe Holding Ag Latch and lockset system
AU671117B2 (en) 1992-06-15 1996-08-15 Scios Inc. Glucagon-like peptide and insulinotropin derivatives
US5686268A (en) * 1992-06-19 1997-11-11 Pfizer Inc. Fused proteins
US5505931A (en) 1993-03-04 1996-04-09 The Dow Chemical Company Acid cleavable compounds, their preparation and use as bifunctional acid-labile crosslinking agents
FR2694294B1 (en) * 1992-07-30 1994-09-09 Rhone Poulenc Rorer Sa Yeast promoter and its user.
NZ255158A (en) 1992-07-31 1997-09-22 Genentech Inc Human growth hormone; stable pharmaceutically acceptable aqueous formulation and means for preparing and storing it
US5602307A (en) 1992-08-12 1997-02-11 Baylor College Of Medicine Non-human animal having predefined allele of a cellular adhesion gene
DE4244915C2 (en) * 1992-08-14 1998-12-03 Widmar Prof Dr Tanner Fungal cells contg. mutated DPM2 mannosyl transferase gene
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5728553A (en) * 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
US5731490A (en) 1992-09-29 1998-03-24 The Ontario Cancer Institute Mutant mouse lacking the expression of interferon regulatory factor 1 (IRF-1)
AU5171293A (en) 1992-10-14 1994-05-09 Regents Of The University Of Colorado, The Ion-pairing of drugs for improved efficacy and delivery
FR2697752B1 (en) * 1992-11-10 1995-04-14 Rhone Poulenc Rorer Sa Antitumor compositions containing taxane derivatives.
TW402639B (en) 1992-12-03 2000-08-21 Transkaryotic Therapies Inc Protein production and protein delivery
US5478745A (en) 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
US5441050A (en) 1992-12-18 1995-08-15 Neoprobe Corporation Radiation responsive surgical instrument
US6221958B1 (en) * 1993-01-06 2001-04-24 Societe De Conseils De Recherches Et D'applications Scientifiques, Sas Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
US5489743A (en) 1993-01-19 1996-02-06 Amgen Inc. Transgenic animal models for thrombocytopenia
US5593972A (en) 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
US5441734A (en) 1993-02-25 1995-08-15 Schering Corporation Metal-interferon-alpha crystals
US5780021A (en) 1993-03-05 1998-07-14 Georgetown University Method for treating type 1 diabetes using α-interferon and/or β-i
HU225496B1 (en) 1993-04-07 2007-01-29 Scios Inc Pharmaceutical compositions of prolonged delivery, containing peptides
US6284727B1 (en) 1993-04-07 2001-09-04 Scios, Inc. Prolonged delivery of peptides
WO1994024160A2 (en) 1993-04-21 1994-10-27 Brigham And Women's Hospital Erythropoietin muteins with enhanced activity
GB9308581D0 (en) 1993-04-26 1993-06-09 Univ Manitoba A method for induction of antigen-specific suppression of immune-response
ATE301201T1 (en) 1993-06-07 2005-08-15 Vical Inc PLASMIDS USABLE FOR GENE THERAPY
DK82893D0 (en) * 1993-07-08 1993-07-08 Novo Nordisk As PEPTIDE
US5512459A (en) 1993-07-20 1996-04-30 Bionebraska, Inc. Enzymatic method for modification or recombinant polypeptides
RU2153332C2 (en) 1993-08-09 2000-07-27 Орион Корпорейшн Method of sensitization of cancer cells to lysis mediated by killer-cells
IL192290A0 (en) 1993-08-17 2008-12-29 Kirin Amgen Inc Erythropoietin analogs
AU7531094A (en) 1993-08-24 1995-03-21 Novo Nordisk A/S Protracted glp-1
GB9317618D0 (en) 1993-08-24 1993-10-06 Royal Free Hosp School Med Polymer modifications
US5521086A (en) 1993-09-16 1996-05-28 Cephalon, Inc. Secretion sequence for the production of a heterologous protein in yeast
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5459031A (en) 1993-11-05 1995-10-17 Amgen Inc. Methods for controlling sialic acid derivatives in recombinant glycoproteins
JPH09506262A (en) 1993-12-08 1997-06-24 ジェンザイム・コーポレイション Method for producing specific antibody
CA2137206A1 (en) 1993-12-09 1995-06-10 John A. Galloway Glucagon-like insulinotropic peptides, compositions and methods
US5705483A (en) 1993-12-09 1998-01-06 Eli Lilly And Company Glucagon-like insulinotropic peptides, compositions and methods
AU1272295A (en) 1993-12-17 1995-07-03 Novo Nordisk A/S Proinsulin-like compounds
DK144093D0 (en) 1993-12-23 1993-12-23 Novo Nordisk As
DK1231268T3 (en) 1994-01-31 2005-11-21 Univ Boston Polyclonal antibody libraries
GB9404270D0 (en) 1994-03-05 1994-04-20 Delta Biotechnology Ltd Yeast strains and modified albumins
US6608182B1 (en) 1994-03-08 2003-08-19 Human Genome Sciences, Inc. Human vascular endothelial growth factor 2
US5629286A (en) * 1994-03-31 1997-05-13 Brewitt; Barbara Homeopathic dilutions of growth factors
US5652352A (en) 1994-03-31 1997-07-29 Amgen Inc. Afamin: a human serum albumin-like gene
US5646113A (en) 1994-04-07 1997-07-08 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
FR2719593B1 (en) 1994-05-06 1996-05-31 Rhone Poulenc Rorer Sa New biologically active polypeptides, their preparation and pharmaceutical composition containing them.
GB9409496D0 (en) 1994-05-12 1994-06-29 London Health Ass Method for improving glycaemic control in diabetes
GB9411356D0 (en) 1994-06-07 1994-07-27 Delta Biotechnology Ltd Yeast strains
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
US5985660A (en) 1994-06-15 1999-11-16 Systemix, Inc. Method of identifying biological response modifiers involved in dendritic and/or lymphoid progenitor cell proliferation and/or differentiation
US5639642A (en) 1994-06-16 1997-06-17 Novo Nordisk A/S Synthetic leader peptide sequences
US5623054A (en) 1994-06-23 1997-04-22 The General Hospital Corporation Crucifer AFT proteins and uses thereof
NZ290089A (en) 1994-07-27 1999-05-28 Queensland Inst Med Res Recombinant polyepitope cytotoxic t lymphocyte (ctl) vaccines
JPH0853500A (en) 1994-08-11 1996-02-27 Asahi Glass Co Ltd Fused protein and gene coding said protein
JPH0851982A (en) 1994-08-11 1996-02-27 Asahi Glass Co Ltd Modified gene coding human serum albumen
JPH0859509A (en) 1994-08-16 1996-03-05 Teijin Ltd Agent for treating cytomegalovirus antigenemia and agent for treating cytomegalovirus infectious disease
US5574008A (en) 1994-08-30 1996-11-12 Eli Lilly And Company Biologically active fragments of glucagon-like insulinotropic peptide
US6071923A (en) 1994-09-16 2000-06-06 Bar-Ilan University Retinoyloxy aryl-substituted alkylene butyrates useful for the treatment of cancer and other proliferative diseases
US5512549A (en) * 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
FR2726576B1 (en) 1994-11-07 1997-01-31 Pf Medicament PRODUCTION OF HYDROPHOBIC PEPTIDE-LIKE PEPTIDES, RECOMBINANT PEPTIDE, CORRESPONDING DNA SEQUENCE
FR2726471B1 (en) 1994-11-07 1997-01-31 Pf Medicament PROCESS FOR IMPROVING THE IMMUNOGENICITY OF AN IMMUNOGENIC COMPOUND OR A HAPTENA AND APPLICATION TO THE PREPARATION OF VACCINES
ES2285701T3 (en) 1994-11-07 2007-11-16 Human Genome Sciences, Inc. TUMOR NECROSIS FACTOR-GAMMA.
AT403167B (en) 1994-11-14 1997-11-25 Immuno Ag SELECTION AND EXPRESSION OF FOREIGN PROTEINS BY MEANS OF A SELECTION-AMPLIFICATION SYSTEM
JPH10510155A (en) 1994-12-07 1998-10-06 バイオネブラスカ・インコーポレイテッド Method for producing peptide using recombinant fusion protein construct
AU4415796A (en) 1994-12-07 1996-06-26 Bionebraska, Inc. Production of c-terminal amidated peptides from recombinant protein constructs
EP1621206A1 (en) * 1994-12-12 2006-02-01 Beth Israel Deaconess Medical Center, Inc. Chimeric cytokines and uses thereof
WO1996020005A1 (en) 1994-12-23 1996-07-04 Novo Nordisk A/S Protracted glp-1 compositions
US20010006943A1 (en) 1994-12-23 2001-07-05 Ejvind Jensen Protracted GLP-1 compositions
US5652224A (en) 1995-02-24 1997-07-29 The Trustees Of The University Of Pennsylvania Methods and compositions for gene therapy for the treatment of defects in lipoprotein metabolism
US5928939A (en) 1995-03-01 1999-07-27 Ludwig Institute For Cancer Research Vascular endothelial growth factor-b and dna coding therefor
JP2758154B2 (en) 1995-04-06 1998-05-28 エフ・ホフマン−ラ ロシユ アーゲー Liquid preparations containing interferon
US6184201B1 (en) 1995-04-14 2001-02-06 Nps Allelix Corp. Intestinotrophic glucagon-like peptide-2 analogs
EP0822830B1 (en) 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
AU2363695A (en) 1995-04-27 1996-11-18 Human Genome Sciences, Inc. Human tumor necrosis factor receptors
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
AU5724996A (en) 1995-05-05 1996-11-21 Eli Lilly And Company Single chain insulin with high bioactivity
US5728915A (en) 1995-05-08 1998-03-17 Children's Hospital, Inc. Transgenic mice which express simian SV 40 T-antigen under control of the retinoblastoma gene promoter
US6387365B1 (en) * 1995-05-19 2002-05-14 Schering Corporation Combination therapy for chronic hepatitis C infection
US5741815A (en) 1995-06-02 1998-04-21 Lai; Ching-San Methods for in vivo reduction of nitric oxide levels and compositions useful therefor
US5804162A (en) 1995-06-07 1998-09-08 Alliance Pharmaceutical Corp. Gas emulsions stabilized with fluorinated ethers having low Ostwald coefficients
US5728707A (en) * 1995-07-21 1998-03-17 Constantia Gruppe Treatment and prevention of primary and metastatic neoplasms with salts of aminoimidazole carboxamide
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
US5840542A (en) 1995-07-28 1998-11-24 Mogam Biotechnology Research Institute Method for manufacture of proinsulin with high export yield
JPH0940699A (en) * 1995-07-28 1997-02-10 Sanwa Kagaku Kenkyusho Co Ltd Polypeptide and its use
DE19530865A1 (en) 1995-08-22 1997-02-27 Michael Dr Med Nauck Active ingredient and agent for parenteral nutrition
JPH0995455A (en) 1995-09-29 1997-04-08 Sumitomo Pharmaceut Co Ltd Renal function improving agent
US5766620A (en) 1995-10-23 1998-06-16 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US5849322A (en) 1995-10-23 1998-12-15 Theratech, Inc. Compositions and methods for buccal delivery of pharmaceutical agents
US5702717A (en) 1995-10-25 1997-12-30 Macromed, Inc. Thermosensitive biodegradable polymers based on poly(ether-ester)block copolymers
PT858343E (en) 1995-11-02 2004-07-30 Schering Corp THERAPY FOR CONTINUOUS INFUSE OF A LOW DOSE OF CYTOKINE
US6048964A (en) 1995-12-12 2000-04-11 Stryker Corporation Compositions and therapeutic methods using morphogenic proteins and stimulatory factors
JP3533857B2 (en) * 1995-12-25 2004-05-31 富士レビオ株式会社 Treponema pallidum fusion antigen and method for measuring anti-treponema pallidum antibody using the fusion antigen
GB9526733D0 (en) * 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US6087129A (en) 1996-01-19 2000-07-11 Betagene, Inc. Recombinant expression of proteins from secretory cell lines
US6110707A (en) 1996-01-19 2000-08-29 Board Of Regents, The University Of Texas System Recombinant expression of proteins from secretory cell lines
CA2246431A1 (en) 1996-01-19 1997-07-24 Board Of Regents, The University Of Texas System Recombinant expression of proteins from secretory cell lines
US6150337A (en) 1996-01-23 2000-11-21 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by Ribavirin in activated T-lymphocytes
US5767097A (en) * 1996-01-23 1998-06-16 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by ribavirin in activated T-lymphocytes
JP2978435B2 (en) 1996-01-24 1999-11-15 チッソ株式会社 Method for producing acryloxypropyl silane
US5616724A (en) 1996-02-21 1997-04-01 Cephalon, Inc. Fused pyrrolo[2,3-c]carbazole-6-ones
DE19639601A1 (en) 1996-02-28 1997-09-04 Bayer Ag Parapox viruses that contain foreign DNA, their production and their use in vaccines
US6045788A (en) * 1996-02-28 2000-04-04 Cornell Research Foundation, Inc. Method of stimulation of immune response with low doses of IL-2
US5912229A (en) 1996-03-01 1999-06-15 Novo Nordisk Als Use of a pharmaceutical composition comprising an appetite-suppressing peptide
ES2187756T3 (en) 1996-03-01 2003-06-16 Novo Nordisk As USE OF A PHARMACEUTICAL COMPOSITION THAT INCLUDES AN APPETIZER SUPPRESSING PEPTIDE.
JP3794748B2 (en) * 1996-03-04 2006-07-12 第一アスビオファーマ株式会社 Method for culturing microorganisms with methanol metabolism
WO1997033904A1 (en) 1996-03-12 1997-09-18 Human Genome Sciences, Inc. Death domain containing receptors
ES2200153T3 (en) 1996-03-13 2004-03-01 Delta Biotechnology Limited FERMENTATION CONTROL.
WO1997033899A1 (en) 1996-03-14 1997-09-18 Human Genome Sciences, Inc. Apoptosis inducing molecule i
CA2248136A1 (en) 1996-03-21 1997-09-25 Human Genome Sciences, Inc. Human endometrial specific steroid-binding factor i, ii and iii
CA2248868A1 (en) 1996-03-22 1997-09-25 Human Genome Sciences, Inc. Apoptosis inducing molecule ii
AU2667197A (en) 1996-04-16 1997-11-07 University Of Miami Stabilized preparations of human troponins and modifications thereof, diagnostic assay methods and assay kits
US5919815A (en) 1996-05-22 1999-07-06 Neuromedica, Inc. Taxane compounds and compositions
JP4172726B2 (en) 1996-05-22 2008-10-29 ルイトポルド・ファーマシューティカルズ・インコーポレーテッド Formulation containing a covalent complex of cis-docosahexaenoic acid and docetaxel
US6300065B1 (en) * 1996-05-31 2001-10-09 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
ES2150832B1 (en) 1996-06-12 2001-06-16 Fichtel & Sachs Ag OPERATING DEVICE FOR THE OPERATION, IN PARTICULAR PNEUMATIC OPERATION, OF A FRICTION CLUTCH.
US6110891A (en) 1996-06-21 2000-08-29 Alizyme Therapeutics Ltd. Lectin compositions and uses thereof
GB2318352A (en) 1996-06-25 1998-04-22 Dejan Markovic Polypeptides mimicking the activity of human erythropoietin
GB9613858D0 (en) 1996-07-02 1996-09-04 Cortecs Ltd Hydrophobic preparations
US5932547A (en) 1996-07-03 1999-08-03 Alza Corporation Non-aqueous polar aprotic peptide formulations
US6110703A (en) 1996-07-05 2000-08-29 Novo Nordisk A/S Method for the production of polypeptides
ES2242227T5 (en) 1996-07-15 2011-12-09 Chugai Seiyaku Kabushiki Kaisha NEW VEGF TYPE FACTOR.
AR008077A1 (en) 1996-07-26 1999-12-09 Talarico Salinas Laura Beatriz A FUSION POLYPEPTIDE OR A SALT OF THE SAME, ITS USE, A PROCESS TO PREPARE THEM, A PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS THEM, AND A VECTOR.
US5959075A (en) * 1996-08-02 1999-09-28 Zymogenetics, Inc. Testis-specific insulin homolog polypeptides
AU4055697A (en) 1996-08-16 1998-03-06 Schering Corporation Mammalian cell surface antigens; related reagents
JP2000516474A (en) 1996-08-16 2000-12-12 ヒューマン ジノーム サイエンシーズ,インコーポレイテッド Human endokine alpha
ATE434034T1 (en) 1996-08-23 2009-07-15 Vegenics Ltd RECOMBINANT VASCULAR ENDOTHELIAL CELL GROWTH FACTOR D (VEGF-D)
US6458924B2 (en) 1996-08-30 2002-10-01 Novo Nordisk A/S Derivatives of GLP-1 analogs
US6006753A (en) 1996-08-30 1999-12-28 Eli Lilly And Company Use of GLP-1 or analogs to abolish catabolic changes after surgery
US6277819B1 (en) 1996-08-30 2001-08-21 Eli Lilly And Company Use of GLP-1 or analogs in treatment of myocardial infarction
US5922761A (en) 1996-09-06 1999-07-13 Medinox, Inc. Methods for in vivo reduction of iron levels and compositions useful therefor
AU4199597A (en) 1996-09-09 1998-04-02 Novo Nordisk A/S A cdna and peptide with relation to cancer and weight loss
US20020052309A1 (en) 1996-09-11 2002-05-02 Athanasius A. Anagnostou Method of treating endothelial injury
WO1998012344A1 (en) 1996-09-18 1998-03-26 Human Genome Sciences, Inc. Human tumor necrosis factor receptor-like genes
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
US5955508A (en) * 1996-10-15 1999-09-21 Loyola University Of Chicago Method for the enhancement of lymphocyte activity against opportunistic microbial pathogens
WO1998017281A1 (en) 1996-10-24 1998-04-30 Vion Pharmaceuticals, Inc. MONOPHOSPHATE PRODRUGS OF β-L-FD4C AND β-L-FddC AS POTENT ANTIVIRAL AGENTS
JP2001503263A (en) 1996-10-25 2001-03-13 ヒューマン ジノーム サイエンシーズ,インコーポレイテッド Neutrokine α
US5908830A (en) 1996-10-31 1999-06-01 Merck & Co., Inc. Combination therapy for the treatment of diabetes and obesity
EP1306092A3 (en) 1996-11-05 2003-05-07 Eli Lilly & Company Use of GLP-1 and analogs administered peripherally, in regulation of obesity
UA65549C2 (en) * 1996-11-05 2004-04-15 Елі Ліллі Енд Компані Use of glucagon-like peptides such as glp-1, glp-1 analog, or glp-1 derivative in methods and compositions for reducing body weight
ATE366584T1 (en) 1996-11-12 2007-08-15 Novo Nordisk As USE OF GLP-1 PEPTIDES
ES2301183T3 (en) 1996-12-03 2008-06-16 Amgen Fremont Inc. COMPLETELY HUMAN ANTIBODY THAT JOINS THE EGFR RECEIVER.
US6130248A (en) 1996-12-30 2000-10-10 Bar-Ilan University Tricarboxylic acid-containing oxyalkyl esters and uses thereof
US5833994A (en) 1997-01-08 1998-11-10 Paracelsian, Inc. Use of the AH receptor and AH receptor ligands to treat or prevent cytopathicity of viral infection
KR20000070174A (en) 1997-01-14 2000-11-25 벤슨 로버트 에이치. Tumor necrosis factor receptors 6α and 6β
JP2001505060A (en) 1997-01-14 2001-04-17 ヒューマン ジノーム サイエンシーズ,インコーポレイテッド Tumor necrosis factor receptor 5
WO1998032867A1 (en) 1997-01-24 1998-07-30 Novo Nordisk A/S Synthetic leader peptide sequences
JP4450870B2 (en) 1997-01-28 2010-04-14 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Binding to death domain containing receptor 4 (DR4: death receptor 4), a member of the TNF-receptor superfamily and TRAIL
EP0921817B1 (en) 1997-01-29 2001-03-28 PolyMASC Pharmaceuticals plc Pegylation process
WO1998036085A1 (en) 1997-02-13 1998-08-20 Applied Phytologics, Inc. Production of mature proteins in plants
US7026447B2 (en) * 1997-10-09 2006-04-11 Human Genome Sciences, Inc. 53 human secreted proteins
US6110970A (en) 1997-03-11 2000-08-29 Beacon Laboratories, Inc. Nitrogen-containing oxyalkylene esters and uses thereof
US6124495A (en) 1997-03-11 2000-09-26 Beacon Laboratories, Inc. Unsaturated oxyalkylene esters and uses thereof
US6110955A (en) 1997-03-11 2000-08-29 Beacon Laboratories, Inc. Metabolically stabilized oxyalkylene esters and uses thereof
US5939455A (en) 1997-03-11 1999-08-17 Beacon Laboratories, Inc. Therapeutic augmentation of oxyalkylene diesters and butyric acid derivatives
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds and uses thereof
US5846774A (en) 1997-03-21 1998-12-08 Bionebraska, Inc. Chlorella virus promoters
US6627729B1 (en) * 1997-03-24 2003-09-30 Zymogenetics, Inc. TML peptides
JP2002510193A (en) 1997-03-31 2002-04-02 イーライ・リリー・アンド・カンパニー Glucagon-like peptide-1 analog
SI0970126T1 (en) 1997-04-14 2001-08-31 Micromet Ag Novel method for the production of antihuman antigen receptors and uses thereof
TW577759B (en) 1997-04-18 2004-03-01 Ipsen Pharma Biotech Sustained release compositions in the form of microcapsules or implants and the process for their preparation
AU7273598A (en) 1997-05-05 1998-11-27 Regents Of The University Of California, The Naphthols useful in antiviral methods
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
CA2292790A1 (en) 1997-05-30 1998-12-03 Human Genome Sciences, Inc. Human tumor necrosis factor receptor tr10
US6071743A (en) 1997-06-02 2000-06-06 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
AU7830398A (en) 1997-06-11 1998-12-30 Human Genome Sciences, Inc. Human tumor necrosis factor receptor tr9
GB9713412D0 (en) 1997-06-26 1997-08-27 Delta Biotechnology Ltd Improved protein expression strains
ATE255634T1 (en) 1997-09-01 2003-12-15 Aventis Pharma Gmbh RECOMBINANT HUMAN ERYTHROPOIETIN WITH ADVANTAGEOUS GLYCOSYLATION PATTERN
JP3201987B2 (en) 1997-09-09 2001-08-27 株式会社オーレック Mowing equipment
HUP9701554D0 (en) 1997-09-18 1997-11-28 Human Oltoanyagtermeloe Gyogys Pharmaceutical composition containing plazma proteins
US6472373B1 (en) 1997-09-21 2002-10-29 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in antiviral treatment naive patients having chronic hepatitis C infection
US6172046B1 (en) * 1997-09-21 2001-01-09 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic Hepatitis C infection
EP1136075B1 (en) 1997-09-21 2003-01-15 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic hepatitis C infection
ES2181281T5 (en) 1997-09-23 2008-04-16 Rentschler Biotechnologie Gmbh BETA INTERFER LIQUID FORMULATIONS.
US6117949A (en) 1998-10-01 2000-09-12 Macromed, Inc. Biodegradable low molecular weight triblock poly (lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6201072B1 (en) * 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
CA2305690A1 (en) 1997-10-09 1999-04-22 Human Genome Sciences, Inc. 53 human secreted proteins
CN1174993C (en) 1997-11-03 2004-11-10 人体基因组科学有限公司 VEGI, an inhibitor of angiogenesis and tumor growth
ES2523599T3 (en) 1997-12-03 2014-11-27 Roche Diagnostics Gmbh Erythropoietin of great specific activity
CA2312190A1 (en) 1997-12-05 1999-06-17 Eli Lilly And Company Glp-1 formulations
WO1999029732A2 (en) 1997-12-08 1999-06-17 Lexigen Pharmaceuticals Corporation Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
AU738804B2 (en) * 1997-12-08 2001-09-27 Bristol-Myers Squibb Company Novel salts of metformin and method
US6380357B2 (en) 1997-12-16 2002-04-30 Eli Lilly And Company Glucagon-like peptide-1 crystals
KR100253916B1 (en) 1997-12-29 2000-05-01 김충환 A process for preparing human proinsulin
US5951996A (en) 1998-02-04 1999-09-14 Czeizler Zaharia; Veronica L. Treatment of chronic diffuse GI bleeding with erythropoietin
US6017545A (en) * 1998-02-10 2000-01-25 Modi; Pankaj Mixed micellar delivery system and method of preparation
US6221378B1 (en) * 1998-02-10 2001-04-24 Generex Pharmaceuticals Incorporated Mixed micellar delivery system and method of preparation
ATE366115T1 (en) 1998-02-13 2007-07-15 Amylin Pharmaceuticals Inc INOTROPIC AND DIURETIC EFFECTS OF EXENDIN AND GLP-1
AU2610699A (en) 1998-02-27 1999-09-15 Novo Nordisk A/S Derivatives of glp-1 analogs
WO1999043705A1 (en) * 1998-02-27 1999-09-02 Novo Nordisk A/S N-terminally truncated glp-1 derivatives
WO1999047160A1 (en) 1998-03-13 1999-09-23 Novo Nordisk A/S Stabilized aqueous peptide solutions
AU3087599A (en) 1998-03-19 1999-10-11 Bionebraska, Inc. Human appetite control by glucagon-like peptide receptor binding compounds
DE19813802A1 (en) 1998-03-27 1999-11-11 Retro Tech Gmbh Anti-viral effects of propolis through inhibition of viral nucleic acid polymerases
WO1999053064A2 (en) 1998-04-13 1999-10-21 Modex Therapeutiques, S.A. Methods of delivering glp-1
CA2270320A1 (en) 1998-04-28 1999-10-28 Patricia Lee Brubaker Novel microsphere composition
US6251868B1 (en) 1998-04-30 2001-06-26 Teijin Limited Method for treating a human immunodeficiency virus infection
CN1119352C (en) 1998-05-15 2003-08-27 中国科学院上海生物化学研究所 Express and purification of human serum albumin in pichia
TWI277424B (en) 1998-05-15 2007-04-01 Schering Corp Combination therapy for eradicating detectable NCV-RNA in antiviral treatment naive patients having chronic hepatitis C infection
SE9802080D0 (en) 1998-06-11 1998-06-11 Hellstroem Pharmaceutical composition for the treatment of functional dyspepsia and / or irritable bowel syndrome and new use of substances therein
SK19042000A3 (en) 1998-06-12 2001-11-06 Bionebraska, Inc. Glucagon-like peptide-1 improves beta-cell response to glucose in subjects with impaired glucose tolerance
CA2330527A1 (en) * 1998-06-15 1999-12-23 Genzyme Transgenics Corporation Erythropoietin analog-human serum albumin fusion
CN1105727C (en) 1998-06-17 2003-04-16 上海海济医药生物工程有限公司 Process for preparing recombined human serum albumin
JP2002518452A (en) 1998-06-24 2002-06-25 エモリ ユニバーシティ Use of 3'-azido-2 ', 3'-dideoxyuridine in combination with other anti-HIV drugs for the manufacture of a therapeutic drug for HIV
EP1091979B1 (en) 1998-06-30 2006-09-20 Novo Nordisk A/S Seeding crystals for the preparation of peptides or proteins
WO2000004171A1 (en) 1998-07-15 2000-01-27 Wisconsin Alumni Research Foundation Treatment of diabetes with synthetic beta cells
AU5027299A (en) 1998-07-31 2000-02-28 Novo Nordisk A/S Use of glp-1 and analogues for preventing type ii diabetes
GB9817084D0 (en) 1998-08-06 1998-10-07 Wood Christopher B A method for promoting extra-heptic production of proteins for the correction of hypoalbuminaemia,anaemia,thrombocytopenia and/or coagulation disorders
ES2335066T3 (en) 1998-08-10 2010-03-18 The Government Of The Usa, As Represented By The Secretary, Department Of Health And Human Services DIFFERENTIATION OF NON-INSULIN CELL PRODUCTS IN GLP-1 OR EXENDIN-4 INSULIN CELLS AND USING THEMSELVES.
US6346543B1 (en) * 1998-08-17 2002-02-12 Aventis Pharma S.A. Use of a taxoid to treat abnormal cell proliferation in the brain
KR20010073033A (en) 1998-08-28 2001-07-31 피터 지. 스트링거 Method for Administering Insulinotropic Peptides
MY155270A (en) 1998-09-24 2015-09-30 Lilly Co Eli Use of glp-1 or analogs in treatment of stroke
US6193997B1 (en) * 1998-09-27 2001-02-27 Generex Pharmaceuticals Inc. Proteinic drug delivery system using membrane mimetics
US6290987B1 (en) 1998-09-27 2001-09-18 Generex Pharmaceuticals, Inc. Mixed liposome pharmaceutical formulation with amphiphiles and phospholipids
US6284725B1 (en) 1998-10-08 2001-09-04 Bionebraska, Inc. Metabolic intervention with GLP-1 to improve the function of ischemic and reperfused tissue
US7259136B2 (en) 1999-04-30 2007-08-21 Amylin Pharmaceuticals, Inc. Compositions and methods for treating peripheral vascular disease
US6429197B1 (en) 1998-10-08 2002-08-06 Bionebraska, Inc. Metabolic intervention with GLP-1 or its biologically active analogues to improve the function of the ischemic and reperfused brain
AU772460B2 (en) 1998-10-16 2004-04-29 Immunex Corporation Inhibitors of platelet activation and recruitment
ES2347884T3 (en) 1998-10-23 2010-11-22 Amgen Inc. METHOD AND COMPOSITIONS FOR THE PREVENTION AND TREATMENT OF THE ANEMIA.
CA2348822A1 (en) 1998-10-30 2000-05-11 Novozymes A/S Glycosylated proteins having reduced allergenicity
EP1135493A2 (en) 1998-11-30 2001-09-26 Eli Lilly And Company Erythropoietic compounds
US6048891A (en) 1998-12-17 2000-04-11 Loma Linda University Medical Center Use of γ-tocopherol and its oxidative metabolite LLU-α in the treatment of natriuretic disease
US6294153B1 (en) 1998-12-21 2001-09-25 Generex Pharmaceuticals, Inc. Aerosol pharmaceutical formulation for pulmonary and nasal delivery
US6312665B1 (en) 1998-12-21 2001-11-06 Generex Pharmaceuticals Incorporated Aerosol formulations for buccal and pulmonary application
US6271200B1 (en) 1998-12-21 2001-08-07 Generex Pharmaceuticals Inc. Proteinic drug delivery system using aerosolized membrane-mimetic amphiphiles
EP1140148B1 (en) 1998-12-22 2005-10-26 Eli Lilly And Company Shelf-stable solution formulation of glucagon-like peptide-1
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
WO2000062759A1 (en) 1999-04-16 2000-10-26 Novo Nordisk A/S Dry, mouldable drug formulation
US6287588B1 (en) 1999-04-29 2001-09-11 Macromed, Inc. Agent delivering system comprised of microparticle and biodegradable gel with an improved releasing profile and methods of use thereof
DE19921537A1 (en) 1999-05-11 2000-11-23 Dieter Hoersch Treating carbohydrate metabolism disorders, especially diabetes, comprises activating insulin-secreting b-cells using glucagon-related peptide, glucose-dependent insulinotropic polypeptide, exendin-4 or related drugs
US6348192B1 (en) * 1999-05-11 2002-02-19 Bayer Corporation Interleukin-2 mutein expressed from mammalian cells
US6514500B1 (en) 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
JP4115671B2 (en) * 1999-05-17 2008-07-09 コンジュケム バイオテクノロジーズ インコーポレイテッド Long-lasting insulinotropic peptides
AU777963B2 (en) 1999-05-19 2004-11-04 Merck Patent Gmbh Expression and export of interferon-alpha proteins as Fc fusion proteins
US6344180B1 (en) 1999-06-15 2002-02-05 Bionebraska, Inc. GLP-1 as a diagnostic test to determine β-cell function and the presence of the condition of IGT and type II diabetes
PT1133312E (en) 1999-06-21 2007-11-30 Lilly Co Eli Synergistic use of thiazolidinediones with glucagon-like peptide-1 and agonists thereof to treat non-insulin dependent diabetes
PE20010288A1 (en) 1999-07-02 2001-03-07 Hoffmann La Roche ERYTHROPOYETIN DERIVATIVES
CA2376641A1 (en) 1999-07-19 2001-01-25 Aaron Morris Chimeric polypeptides of serum albumin and uses related thereto
US20020048571A1 (en) * 1999-07-19 2002-04-25 Jeno Gyuris Chimeric polypeptides of serum albumin and uses related thereto
US7144574B2 (en) * 1999-08-27 2006-12-05 Maxygen Aps Interferon β variants and conjugates
TWI260321B (en) 1999-09-22 2006-08-21 Bristol Myers Squibb Co Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
PH12000002657B1 (en) 1999-10-12 2006-02-21 Bristol Myers Squibb Co C-aryl glucoside SGLT2 inhibitors
WO2001029242A2 (en) 1999-10-21 2001-04-26 Monsanto Company Post-translational modification of recombinant proteins produced in plants
WO2001030320A1 (en) 1999-10-22 2001-05-03 Amgen Inc. Biodegradable microparticles with novel erythropoietin stimulating protein
DE60037535D1 (en) 1999-11-03 2008-01-31 Novo Nordisk As USE OF A GROWTH HORMONE OR A GROWTH HORMONE SECRETION THROUGH MEANS OF APPETITE SUPPRESSION OR SATURATION INDUCTION
CA2324801A1 (en) 1999-11-10 2001-05-10 Andrew Gordon Swick Use of apo b secretion/mtp inhibitors and anti-obesity agents
AU2154401A (en) 1999-11-12 2001-05-30 Merck Patent Gmbh Erythropoietin forms with improved properties
US6569832B1 (en) * 1999-11-12 2003-05-27 Novo Nordisk A/S Inhibition of beta cell degeneration
US6866843B2 (en) 1999-12-06 2005-03-15 Viacell, Inc. Method of transplanting in a mammal and treating diabetes mellitus by administering a pseudo-islet like aggregate differentiated from a nestin-positive pancreatic stem cell
US20020025306A1 (en) 2000-01-07 2002-02-28 Baetge Edward E. Methods of increasing the glucose responsiveness of pancreatic ss-cells
EP1125579A3 (en) 2000-01-18 2003-01-02 Pfizer Products Inc. Uses of agrp-melanocortin receptor binding modulating compounds
PT1257577E (en) 2000-01-27 2004-08-31 Lilly Co Eli METHOD FOR SOLUBILIZING GLUCAGON TYPE 1 PEPTIDE COMPOUNDS
AU2001228327A1 (en) 2000-01-31 2001-08-14 Novo-Nordisk A/S Crystallisation of a glp-1 analogue
WO2001068112A2 (en) 2000-03-14 2001-09-20 Goeke Burkhard Effects of glucagon-like peptide-1 (7-36) on antro-pyloro-duodenal motility
JP4237375B2 (en) * 2000-03-31 2009-03-11 アスビオファーマ株式会社 Pharmaceutical composition used for treatment or prevention of ischemic disease
US8017357B2 (en) * 2000-04-07 2011-09-13 Eiken Kagaku Kabushiki Kaisha Method of amplifying nucleic acid by using double-stranded nucleic acid as template
US6946134B1 (en) * 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
AU2001264563A1 (en) 2000-04-12 2001-10-30 Human Genome Sciences, Inc. Albumin fusion proteins
US20050100991A1 (en) 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
DK1274728T3 (en) 2000-04-21 2008-09-01 Amgen Inc Methods and compositions for the prevention and treatment of anemia
IL142707A0 (en) 2000-04-27 2002-03-10 Pfizer Prod Inc Methods of treating obesity using a neurotensin receptor ligand
CA2408685C (en) * 2000-05-15 2011-02-01 F. Hoffmann-La Roche Ag Liquid pharmaceutical composition containing an erythropoietin derivate
WO2001087322A2 (en) 2000-05-17 2001-11-22 Bionebraska, Inc. Peptide pharmaceutical formulations
DE60134403D1 (en) 2000-05-19 2008-07-24 Amylin Pharmaceuticals Inc TREATMENT OF ACUTE CORONARY SYNDROME WITH GLP-1
DE60124710T2 (en) 2000-06-16 2007-09-13 Eli Lilly And Co., Indianapolis ANALOG OF GLUCAGON SIMILAR PEPTIDE-1
DK1326630T3 (en) 2000-09-18 2008-09-15 Sanos Bioscience As Use of GLP-2 peptides
DE60142351D1 (en) 2000-10-20 2010-07-22 Amylin Pharmaceuticals Inc TREATMENT OF HYPOACTIVE MYOKARD AND DIABETIC HEART MYOPATHY WITH A GLP-1 PEPTIDE
US7101561B2 (en) * 2000-12-01 2006-09-05 Innogenetics N.V. Purified hepatitis C virus envelope proteins for diagnostic and therapeutic use
WO2002045712A1 (en) 2000-12-04 2002-06-13 Biovitrum Ab Novel method and use
DE60139430D1 (en) * 2000-12-07 2009-09-10 Lilly Co Eli GLP-1 fusion proteins
EP1390061A2 (en) 2000-12-13 2004-02-25 Eli Lilly And Company Chronic treatment regimen using glucagon-like insulinotropic peptides
US7199217B2 (en) 2000-12-13 2007-04-03 Eli Lilly And Company Amidated glucagon-like peptide-1
WO2002066062A2 (en) 2001-02-01 2002-08-29 Drucker Daniel J Enhancement of glp-2 activity
KR100399337B1 (en) 2001-02-07 2003-09-26 드림바이오젠 주식회사 Method for Cell-free Protein Post-translational Modification
JP2004518756A (en) 2001-03-07 2004-06-24 ノボ ノルディスク アクティーゼルスカブ Use of a combination of a GLP-1 analog and a derivative of a PPAR ligand
US20020169128A1 (en) 2001-04-09 2002-11-14 Geroge Sigounas Erythropoietin ameliorates chemotherapy-induced toxicity in vivo
US20060084794A1 (en) * 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
US7507413B2 (en) * 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
US20050054051A1 (en) * 2001-04-12 2005-03-10 Human Genome Sciences, Inc. Albumin fusion proteins
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
WO2002085406A1 (en) 2001-04-24 2002-10-31 Restoragen, Inc. Methods and compositions for treating conditions associated with insulin resistance
EP1401477A4 (en) 2001-05-25 2005-02-02 Human Genome Sciences Chemokine beta-1 fusion proteins
WO2002098348A2 (en) 2001-06-01 2002-12-12 Eli Lilly And Company Glp-1 formulations with protracted time action
WO2003003971A2 (en) 2001-06-20 2003-01-16 Merck Sante Use of antidiabetics for making a medicine with cicatrizing effect
DK1412384T3 (en) 2001-06-28 2008-04-28 Novo Nordisk As Stable formulation of modified GLP-1
CA2455963C (en) 2001-07-31 2017-07-04 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services Glp-1, exendin-4, peptide analogs and uses thereof
JP2005509409A (en) 2001-08-08 2005-04-14 ジェンザイム・コーポレーション Methods for treating diabetes and other glycemic disorders
JP2004537580A (en) 2001-08-10 2004-12-16 エピックス メディカル, インコーポレイテッド Polypeptide conjugates with extended circulating half-life
HUP0501192A3 (en) 2001-08-23 2006-06-28 Lilly Co Eli Glucagon-like peptide-1 analogs
AU2002332041A1 (en) * 2001-10-05 2003-04-22 Human Genome Sciences, Inc. Albumin fusion proteins
EP2042196B1 (en) * 2001-10-10 2016-07-13 ratiopharm GmbH Remodelling and glycoconjugation of Granulocyte Colony Stimulating Factor (G-CSF)
US20080167238A1 (en) * 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
CA2484556A1 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
WO2003054182A2 (en) 2001-12-21 2003-07-03 Nexia Biotechnologies, Inc. Production of butyrylcholinesterases in transgenic mammals
US20080194481A1 (en) 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
EP1997829A1 (en) 2001-12-21 2008-12-03 Human Genome Sciences, Inc. Albumin fusion proteins
US6682190B2 (en) * 2002-01-31 2004-01-27 Hewlett-Packard Development Company, L.P. Controlling media curl in print-zone
AU2003210806A1 (en) 2002-03-05 2003-09-22 Eli Lilly And Company Heterologous g-csf fusion proteins
WO2003084563A1 (en) 2002-04-04 2003-10-16 Novo Nordisk A/S Glp-1 agonist and cardiovascular complications
CN1241946C (en) 2002-07-01 2006-02-15 美国福源集团 Human serum albumins recombined merge protein having hyperplasia stimulation function to multiple cells
JP4211375B2 (en) 2002-12-10 2009-01-21 セイコーエプソン株式会社 Data transfer control method, data transfer circuit device, and printing apparatus having the same
US6913890B2 (en) 2002-12-18 2005-07-05 Palo Alto Research Center Incorporated Process for preparing albumin protein conjugated oligonucleotide probes
CA2513213C (en) 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
KR100758755B1 (en) 2003-06-12 2007-09-14 일라이 릴리 앤드 캄파니 -1 glp-1 analog fusion proteins
PL1729795T3 (en) * 2004-02-09 2016-08-31 Human Genome Sciences Inc Albumin fusion proteins
US20060051859A1 (en) * 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs
WO2006073890A2 (en) 2004-12-24 2006-07-13 Amylin Pharmaceuticals, Inc. Use of glp-1 and agonists thereof to prevent cardiac myocyte apoptosis
DK1888103T3 (en) 2005-04-11 2012-04-23 Amylin Pharmaceuticals Inc USE OF GLP-1, EXENDIN AND AGONISTS THEREOF TO DELAY OR PREVENT CARDIAL REMODELING
KR20080071119A (en) 2005-08-12 2008-08-01 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
US8008257B2 (en) * 2005-10-20 2011-08-30 University Of Ottawa Heart Institute ANF fusion proteins
KR20090031368A (en) 2006-05-26 2009-03-25 아밀린 파마슈티칼스, 인크. Compositions and method for treatment of congestive heart failure
WO2007146038A2 (en) 2006-06-07 2007-12-21 Human Genome Sciences, Inc. Albumin fusion proteins
WO2008019143A2 (en) 2006-08-04 2008-02-14 Amylin Pharmaceuticals, Inc Use of exendins and glp-i receptor agonists for altering lipoprotein particle size and subclass composition

Also Published As

Publication number Publication date
US20060194735A1 (en) 2006-08-31
CA2471363A1 (en) 2003-07-24
US20090099073A1 (en) 2009-04-16
WO2003060071A3 (en) 2004-02-26
US20140303077A1 (en) 2014-10-09
EP1463751B1 (en) 2013-05-22
US8252739B2 (en) 2012-08-28
US20080146503A1 (en) 2008-06-19
JP2013208118A (en) 2013-10-10
EP1997829A1 (en) 2008-12-03
ES2545090T3 (en) 2015-09-08
US20160152687A1 (en) 2016-06-02
ES2425738T3 (en) 2013-10-17
JP5705418B2 (en) 2015-04-22
US7847079B2 (en) 2010-12-07
US20080153751A1 (en) 2008-06-26
EP2277888A2 (en) 2011-01-26
ES2500918T3 (en) 2014-10-01
US7238667B2 (en) 2007-07-03
US8211439B2 (en) 2012-07-03
US9221896B2 (en) 2015-12-29
EP2277910A1 (en) 2011-01-26
JP2009213477A (en) 2009-09-24
US9296809B2 (en) 2016-03-29
US7592010B2 (en) 2009-09-22
US20080161243A1 (en) 2008-07-03
EP2261250A1 (en) 2010-12-15
US20050186664A1 (en) 2005-08-25
US20120046221A1 (en) 2012-02-23
US20150203568A1 (en) 2015-07-23
JP2005514060A (en) 2005-05-19
US20080213886A1 (en) 2008-09-04
JP2013172734A (en) 2013-09-05
US7141547B2 (en) 2006-11-28
EP1463751A4 (en) 2005-03-23
EP2277888A3 (en) 2011-04-27
AU2002364586A8 (en) 2003-07-30
CY2014040I1 (en) 2016-04-13
JP2014043448A (en) 2014-03-13
US20090093402A1 (en) 2009-04-09
CA2841097A1 (en) 2003-07-24
JP2015120714A (en) 2015-07-02
EP2277889A2 (en) 2011-01-26
WO2003060071A2 (en) 2003-07-24
US8071539B2 (en) 2011-12-06
JP5829245B2 (en) 2015-12-09
PT1463751E (en) 2013-08-26
US20140179596A1 (en) 2014-06-26
US20080167239A1 (en) 2008-07-10
US20150329619A1 (en) 2015-11-19
US20170096472A1 (en) 2017-04-06
US8012464B2 (en) 2011-09-06
EP2261250B1 (en) 2015-07-01
US8993517B2 (en) 2015-03-31
DK1463751T3 (en) 2013-08-26
CY2014040I2 (en) 2016-04-13
US20070259815A1 (en) 2007-11-08
US20060276396A1 (en) 2006-12-07
EP2277889A3 (en) 2011-04-27
US20080167240A1 (en) 2008-07-10
JP2006176514A (en) 2006-07-06
EP2990417A1 (en) 2016-03-02
JP5424521B2 (en) 2014-02-26
US20130150296A1 (en) 2013-06-13
US8513189B2 (en) 2013-08-20
CY1114265T1 (en) 2016-04-13
US7799759B2 (en) 2010-09-21
EP1463751A2 (en) 2004-10-06
US20110009312A1 (en) 2011-01-13
US20070244047A1 (en) 2007-10-18
JP2011015690A (en) 2011-01-27
AU2002364586A1 (en) 2003-07-30
JP2015126746A (en) 2015-07-09
EP2277889B1 (en) 2014-07-09

Similar Documents

Publication Publication Date Title
CA2471363C (en) Albumin fusion proteins
US10711051B2 (en) Multivalent heteromultimer scaffold design and constructs
US11248037B2 (en) Multivalent heteromultimer scaffold design and constructs
US20080167238A1 (en) Albumin Fusion Proteins
EP2906237A2 (en) Multivalent heteromultimer scaffold design an constructs

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20210831

MKLA Lapsed

Effective date: 20191223