CA2405550A1 - Albumin fusion proteins - Google Patents

Albumin fusion proteins Download PDF

Info

Publication number
CA2405550A1
CA2405550A1 CA002405550A CA2405550A CA2405550A1 CA 2405550 A1 CA2405550 A1 CA 2405550A1 CA 002405550 A CA002405550 A CA 002405550A CA 2405550 A CA2405550 A CA 2405550A CA 2405550 A1 CA2405550 A1 CA 2405550A1
Authority
CA
Canada
Prior art keywords
albumin
protein
variant
fragment
albumin fusion
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.)
Abandoned
Application number
CA002405550A
Other languages
French (fr)
Inventor
Craig A. Rosen
William A. Haseltine
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
Individual
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=27394014&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2405550(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Publication of CA2405550A1 publication Critical patent/CA2405550A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4846Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • 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
    • A61K38/38Albumins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • 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
    • A61P13/12Drugs 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/18Feminine contraceptives
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/12Keratolytics, e.g. wart or anti-corn preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • A61P19/08Drugs 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • 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
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • 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
    • 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
    • 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
    • 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
    • A61P31/12Antivirals
    • 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
    • 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
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • A61P33/12Schistosomicides
    • 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
    • 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
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • 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
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
    • 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
    • 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
    • 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
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs 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
    • 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
    • C07K14/575Hormones
    • 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
    • C07K14/65Insulin-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
    • C07K14/705Receptors; 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
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • 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
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • C07K2319/75Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor containing a fusion for activation of a cell surface receptor, e.g. thrombopoeitin, NPY and other peptide hormones
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

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 maki ng 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 o f treating, preventing, or ameliorating diseases, disorders or conditions usin g albumin fusion proteins of the invention.

Description

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

~~ TTENANT LES PAGES 214 A 229 NOTE : Pour les tomes additionels, veuillez contacter 1e 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 NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:
ALBUMIN FUSION PROTEINS
BACKGROUND OF THE INVENTION
The invention relates generally to Therapeutic proteins (including, but not limited to, a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or fragments or variants of albumin. The invention further relates to Therapeutic proteins (including, but not limited to, a polypeptide, antibody, or peptide, or fragments and variants - thereof) fused to albumin or fragments or variants of albumin, that exhibit extended. shelf life and/or extended or therapeutic activity in solution. These fusion proteins are herein collectively referred to as "albumin fusion proteins. of the invention." The invention encompasses therapeutic albumin fusion proteins, compositions, pharmaceutical compositions, formulations and kits. 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 using these nucleic acids, vectors, and/or host cells. .
The invention is also directed to methods of in vitro stabilizing a Therapeutic protein via fusion or conjugation of the Therapeutic protein to albumin or fragments or variants of albumin.
Human serum albumin (HSA, or HA), a protein of 585 amino acids in its mature form (as shown in Figure 15 or in SEQ ID N0:18), 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.
The role of albumin as a carrier molecule and its inert nature are desirable properties for use as a carrier andtransporter of polypeptides in vivo. The use of .albumin as -a component of an albumin fusion protein as a carrier for various .proteins has been suggested in WO 93/15199, WO 93/15200, and EP 413 622. The use of N-terminal fragments of HA
for~fusions to polypeptides has also been proposed (EP 399 666). Fusion of albumin to the HA, or a fragment thereof, is joined to the DNA coding for the Therapeutic protein. A
suitable host is then transformed or transfected with the fused nucleotide sequences, so arranged on a suitable plasmid as to express a fusion polypeptide. The expression may be effected ih vitro from, for example, prokaryotic or eukaryotic cells, or in vivo e.g. from a transgenic organism. -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. Many protein and peptide drugs also require the addition of high concentrations .of other protein such as albumin to reduce or prevent loss of protein due to binding to the container.
This is a major concern with respect to proteins such as IFN. For this reason, many Therapeutic proteins are formulated in combination with large proportion of albumin carrier,molecule (100-1000 fold excess), though this is an undesirable and expensive feature of the formulation.
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
The present invention is based, in part, on the discovery that Therapeutic proteins 2S may be stabilized to extend the shelf-life, and/oi to retain the Therapeutic protein's activity for extended periods of time in solution, in vitro andlor in vivo, by genetically or chemically fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to stabilize the protein and/or its activity.
In addition it has been determined that the use of albumin-fusion proteins or albumin conjugated proteins may ' reduce the need to formulate protein solutions with large excesses of carrier proteins (such as albumin, unfused) to prevent loss of Therapeutic proteins due to factors such as binding to the container.
The present invention encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or a fragment (portion) or variant of albumin. The present inventiowalso encompasses.albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or a fragment (portion) or variant of albumin, that is sufficient to prolong the shelf life- of . the Therapeutic protein, andlor stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro andlor in vivo. 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.
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 portion corresponding to a Therapeutic 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.
' In another embodiment, the invention includes a method of extending the shelf life of a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and i S
variants thereof) comprising the step of fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to extend the shelf-life of the Therapeutic protein. In a preferred embodiment, the Therapeutic protein used according to this method is fused to the albumin, or the fragment or variant of albumin. In a most preferred embodiment, the Therapeutic protein used according to this method is fused to albumin, or a' fragment or variant of albumin, via recombinant DNA
technology or genetic engineering.
In another embodiment, the invention includes a method of stabilizing a Therapeutic protein (e.g., a polypeptide, antibody, orpeptide, dr fragments and variants thereof) in solution, comprising the step of fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin,'that is sufficient to stabilize the Therapeutic protein. In a preferred embodiment, the. Therapeutic protein used according to this method is fused to the albumin, or the fragment or variant of albumin. In a most preferred embodiment, the Therapeutic protein used according to this method is fused to albumin, or a fragment or variant of albumin, via recombinant DNA technology or genetic engineering.
The present invention further includes transgenic organisms modified to contain the nucleic acid molecules of the invention, preferably modified to express the albumin fusion proteins encoded by the nucleic acid molecules.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 depicts the extended shelf life of an HA fusion protein in terms of the biological activity (Nb2 cell proliferation) of HA-hGH remaining after incubation in cell culture media for up to. 5 weeks at 37°C. Under these conditions, hGH
has no observed activity by week 2.
Figure 2 depicts the extended shelf-life of an HA fusion protein in terms of the stable biological activity (Nb2 cell proliferation) of HA-hGH remaining after incubation in cell culture media for up to 3 weeks at 4, 37, or 50°C. Data is normalized to the biological activity of hGH at time zero. . .
Figures 3A and 3B compare the biological activity of HA-hGH with hGH in the Nb2 cell proliferation assay. Figure 3A shows proliferation after 24 hours of incubation with various concentrations of hGH or the albumin fusion protein, and Figure 3B shows proliferation after 48 hours of incubation with various concentrations of hGH
or the albumin fusion protein.
Figure 4 shows a map of a plasmid (pPPC0005) that can be used as the, base vector into which polynucleotides encoding the Therapeutic proteins (including polypeptide and fragments and variants thereof) may be cloned to form HA-fusions. Plasmid Map key:
PRBlp: PRBI S: ce~evisiae promoter; FL: Fusion leader sequence; rHA: cDNA
encoding HA: ADHlt: ADHI S. cerevisiae terminator; T3: T3 sequencing primer site; T7:

sequencing primer site; Amp R: (3-lactamase gene; ori: origin of replication.
Please note that in ,the provisional applications to which this application claims priority, the plasmid in Figure 4 was .labeled pPPC0006, instead of pPPC0005. In addition the drawing of this plasmid did not show certain pertinent restriction sites in this vector. Thus in the present application, the drawing is labeled pPPC0005 and more restriction sites of the same vector are shown.
Figure 5 compares the recovery of vial-stored HA-IFN solutions of various concentrations with a stock solution after 48 or 72 hours of storage.
Figure 6 compares the activity of an HA-a,-IFN fusion protein after administration to . monkeys via IV or SC.
Figure 7 describes the bioavailability and stability of an HA-a.-IFN fusion protein.
a WO 01/79442 ~ PCT/USO1/11850 Figure $ is a map of an expression vector for the production of HA-- IFN.
Figure 9 shows the location of loops in HA.
Figure 10 is an example of the modification of an HA loop.
Figure 11 is a representation of the HA loops.
Figure 12 shows the HA loop IV.
Figure 13 shows the tertiary structure of HA.
' Figure 14 shows an example of a scFv-HA fusion Figure 15 shows the amino acid sequence of the mature form of human, albumin (SEQ ID N0:18) and a polynucleotide encoding it (SEQ ID N0:17).
DETAILED DESCRIPTION
As described above, the present invention is based, in part, on the discovery that a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) may be stabilized to extend the shelf-life and/or retain the Therapeutic protein's activity for extended periods of time in solution (or in a pharmaceutical composition) in vitro and%or in vivo, by genetically fusing or chemically conjugating the Therapeutic protein, polypeptide or peptide to all or a portion of albumin sufficient to stabilize the protein and its activity.
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 chemical conjugation to one another: The Therapeutic protein and albumin protein, once part of the albumin fusion protein, may 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 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 andlor 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 WO 01/79442 . PCT/USO1/11850 albumin protein. In preferred embodiments, the serum albumin protein component of the albumin fusion protein is the mature portion 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 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 I
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 protien.
In further embodiments, the . invention provides an albumin fusion protein - comprising, or alternatively consisting of, a biologically active andlor therapeutically active 1 f 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. .
~ Therapeutic proteins As stated 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 or cherizical conjugation.
As used herein, "Therapeutic protein" refers to proteins, polypepiides, 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 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.
Additionally, the term "Therapeutic protein" may refer to the endogenous or naturally occurring correlate of a Therapeutic protein.
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-liriiiting WO 01/79442 . ~ PCT/USO1/11850 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.
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 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.
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 viv~ or in vitro. For example, a desirable effect may be' assayed in cell culture.
As an example, when hGH is the Therapeutic protein, 'the effects of hGH on cell proliferation as described in Example 1 may be used as the endpoint for which therapeutic activity is measured. Such in vitro or cell culture assays are commonly available for many 20. 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 of Table 1.
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 25, -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 Iocations..along the polypeptide backbone. There are usually two.
major types of glycosylation: glycosylation characterized by O-linked oligosaccharides, which are attached 30 to serine or threonine residues; and glycosylation characterized by N-linked oligosaccharides, which are attached to asparagine residues in an Asn-X-Ser/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 35 carbohydrate units within the chains at different glycosylation sites.
Glycosylation isomers are also common at the same site within a given cell type. ' . ,-For example, several types of human interferon are glycosylated. Natural human interferon-a2 is O-glycosylated at threonine 106, and N-glycosylation~occurs at asparagine 72 in interferon-a.14 (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-(31a.
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 unglycosyIated analog (interferon-(31b) engineered with sequence modifcations to enhance stability (Hosoi et al., J. Interferon Res. 8:375 (1988; Karpusas et al., CeII 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., Eur. J. Biochem 242:191 (1996); Sareneva et al,. Biochem J. 303:831 (1994:); .
Sareneva ~ et al., J. Interferon Res: 13:267 ( 1993)). Mixed O-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 O-linked glycosylation occurs at a seririe residue located at position 126 (Lai et al., J. Biol. Chem.
261:3116 (1986); Broudy et al., Arch. Biochem. Biophys. 265:329 (1988)).
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 manipulations) 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.
Therapeutic proteins (particularly those disclosed in Table 1) and their nucleic 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 GenSeq. as shown in Table 1.
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 l, or fragment or variable thereof.
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. The "Therapeutic WO 01/79442 ~ PCT/USO1/11850 Protein .X" column discloses Therapeutic protein molecules followed by parentheses containing scientific and brand names 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 (as defined by the amino acid sequence obtainable from the CAS and Genbank accession numbers), or to the entire group of Therapeutic proteins associated with a given Therapeutic protein molecule disclosed in this colurxin. The "Exemplary Identifier" column provides Chemical Abstracts Services (CAS) Registry Numbers (published by the American Chemical ~ Society) and/or Genbank Accession Numbers ((e.g., Locus ID, NP_XXXXX (Reference Sequence Protein), and XP_XXXXX (Model Protein) identifiers available through the national Center for Biotechnology Information (NCBI) webpage at www.ncbi.nlm.nih.gov) that correspond to entries in the CAS Registry or Genbank database which contain an amino acid sequence of the Therapeutic Protein Molecule or of a fragment or variant of the Therapeutic Protein Molecule. In addition GenSeq Accession numbers andlor journal publication citations axe given to identify the exemplary amino acid sequence for some polypeptides.
The summary pages associated with ~ each of these CAS and Genbank and GenSeq Accession Numbers as well as the cited journal publications (e.g., .PubMed ID
number (PMID)) are each incorporated by reference in their entireties, particularly with respect to the amino acid sequences described therein. The "PCT/Patent Reference" column provides U . S .
Patent numbers, or PCT International Publication Numbers corresponding to patents and/or published patent applications that describe the Therapeutic protein molecule.
Each of the patents and/or published patent applications cited in the "PCT/Patent Reference" column are herein incorporated by reference in their entireties. In particular, the amino acid sequences of the specified pblypeptide set forth in the sequence listing of each cited "PCT/Patent Reference", the variants of these amino acid sequences (mutations, fragments, etc.) set -forth, for example, in the detailed description of each cited "PCT/Patent Reference", the therapeutic indications set forth, for example, in the detailed description of each cited "PCTIPatent Reference", and the activity asssaysfor the specified polypeptide set forth in the detailed description, and more particularly, the examples of each cited "PCT/Patent Reference" are incorporated herein by reference. The "Biological Activity"
column describes Biological activities associated, with the Therapeutic protein molecule. Tlie "Exemplary Activity Assay" column provides references that describe assays which may be used to test the therapeutic and/or biological activity of a Therapeutic protein or an albumin fusion protein of the invention comprising a Therapeutic protein X portion. Each of the references cited in the "Exemplary Activity Assay" column are, herein incorporated by reference in their entireties, particularly with respect to the description of the respective activity assay described in the reference (see Methods section, for example) for . assaying the corresponding biological activity set forth in the "Biological Activity"
column of Table 1.

WO 01/79442 ~ PCT/USO1/11850 The "Preferred Indication Y" column describes disease, disorders, and/or conditions that may be treated, prevented, diagnosed, or ameliorated by Therapeutic protein X
or an albumin fusion protein of the invention comprising a Therapeutic protein X
portion.

o ~

U

U

~U

x .c z ,~ o ~a v ~ U n ~ w n. ~ ' U r .o x ~, err"' ~
a ~U ~ ~

~
N

O

a U N i.U. U y ~ ~ ~ .t~..
> O ~ ~ ~ M L: 'V
a N ~ c~ a3 'C3 ~ U N ~ p ~' ~
A
' ' ~

U ..
~ ) +..
O ~ .fy( Gr O~
O O x C ~O ~ U
.
O\
O O -C
V
Jy ' ~+~cN O
4~ p N ,_, r O ~
~.
'~ T3 ~ I~ O :.O ~ a5 O\
N
~ O

U N U ,~,~ O cn a - ~
> U ~ ' O ~ i-. ~ +~
~'~c~'~o~oo T
~~U~~~0.

, x d , o ~ o o > 'a o G ~ .~ o b .~ m '~
~ ~ ~, a~
~

~ U U
' .~.~4-r O0l,y ~+.. ~
~
"
d C7.,y; O O~ ~ ~ ~ cd ~ .
. h ~ a.., +~ b0 'O >> v~ ~

N ~ Z ~ . ~ O ~ U ~ ~
N "
'~
~
U

..C," -U G1 .~
~ , y -a ..~. r~
.~, O cc5 ~d U G'' U .O CA ~" ~ b Ov ~

4~ , ~ >, O ~ U N
~ ,~ v~ C ~ ~~ ~ O'p a) r.~., ~ ~, ~ O ~ ~ .~ ~ .~ O ~ 'C x ,~
~ cCi O
~

Y
C U U '~ ~ ~ ~
;~ c b0 N ~ U ~ N ~ N O
~
"

w ~, " (~
.-. .-~ ~ N ~ > a) ~ cC a ~-" ~
O C ~ c~ W ~t ' m ~~ c~ ~,br~~~a ~ ~.~wzx ~U~
w U
~ ~

O x ~~,~
O O
.' ' O
N

., U ,~; U
~ F-.
+-~
.r~.
~

U

+~ O U ~ O ~, e~ 1, U ,, '.' ~ ~C ' N.' ~~~
i.., ~~n~'~bA..flV'~Gi ~. ~"

d ~ .
"ow U
o~ >'~ ~ ~ ~ ~ ~= o ~-.~~'x ~ .

~s .~~~U'N
d ~o~~..fla,~r~~'ro~o n.
.~ v .
~

_ p s.c o ' N i ~ ~
' .-, ~ .~ v ~ . 3 ~ ~
~

> p ~ ~~o~ a ~' ~'-o ' o ~cn >, ~
.. ...
~
~

o ~ ~ ~ ~, ~ ~ .~ ~
." 'x x ~s " '~

' ,~ ~ ~ ._.. y ~ ~ U vU7 ~ ~ > ..O
~ ~
~ ~' o ~
~ ' ' a~ -~s w a x a~ w a~o Zs ~
d np.'y ~o~
d~
'~~~~
c ~ .
c ~, .

'J

c I~ ~ N
a ~

N O O O

W

p~~ OOo ~.
U
t~ , ~

Y

H

~f' M CO V7 O ~ v~1 0 w ~ ~
lm z a ~

V p U ~ U U n ~

O ~ O .~

p ~ a ~o u. ~

o~na~c u~ d .
od ~

'b ~ o -~

o .
o s >, o b ~ o .
a ~" o. g ~ ~
~

~ ~ ~o ~Qx a~
~

.

U

U

U
b ~ C~1 a N
Q

a... C
c~ O +~
~

O ~ ~ U
f .
' ~
V
~

~ , N
~
j o, ~ ~ x ;-p "
~r o a~ i o ~
Y

~ a~ ~' ~
L~' ftf U
~

~. ~ ~ , U w U ~" y,U"
~ t-~
Cn ~ Cue'. , .D U (~

w ~.~~~ , UN..c ~s g ~ N ~ .t',~ U ~ O i-.
2y O

ate.' .~ ~ ' ,.C .~ V N~ ~ m ~ .~ ~ 4 N
~

~ w i ~ _~ 3 p'~ ~ ~ .~ ~ ~
a ~ ~on a.
>' 'c~ .
~ ~ -o '~~

x ~

b .-. ~ ~ U N C!~ N
."' ("., N ~ ~ ~ ~ ~
,..U~ ~ ~
O' k O
O

N t" ~' U ~ ~ U ~ ~ ~
' ~ x ~3 v7 -G y ~ ' .C
O O
H
~ .
~
U

N O N .O U ~
m c U ~O ~. ~ ~
a a x O
~ C
~ .~

~ ~
't~,~ N O ~ ~ N O ~ v7 .
' b11 -~~ ~ 3 0 0 ~' . ~ o~E
.~ ~
e o ~ ' ~ 3 , ~
~ ~t ~ ~.' , ~' ~ :_c U N +U.. r~~1 V ~ ~ O
~ N N +.' Y ~ N ~ C'..

> ~ ~ 3 ~ o.~ b ~ ~ .~ ~ ~o i ~ ~ ~ ~ ~

~''UNN~U 2~
~ N
'~
~O~
U' ~
"
N

U

e-! .f Q ~," W ..
U bA.a C.
. i ~
C
r C
.L
~
cC U ~1...-m U ~ SZ, ~'.i," cC cci . CC
tC

N N
~O N

O N

O d' ~ 01 33 w a3 p ~ N

N

O
U

O.b b ~

tiA O O

O cC ~

~ b Q p pp O y U

"
by ~ ~
G

.

o ~
.
~

"
~
U

U.
' , HE~~
c oU

~D
.
' ~

~
',~
+3 ~
-a -a , w ~ ~.
o a~
o E~E-~>'~ G~~~1Q~7.~Ca -o ~

-d . + N o y p .~
by ~..
o o ~
cc o x ~~ ~-03 a;
~
~

~ ~, O +:. _ ~ C/~ ~ O O U ~' ~
~
U

o0 o ~.~~z ~ _ ~~~ ~ ~ ;,~~aM

W~ ;-~ ~ 'O ~
O . ~
~ ~
~ "~'~
~
.b ~
'~
d ~
N
gyp .

c 4.
C .
N
' x ~
' p ~
;
~
~

~ 3 ~ x ~
Uh o ~W, o ' r _ ~, o o ]
~
m.~
U+.
bA~

~ ,...
, .~
o G > 0 0 ~ ~
~n ~
,~
a ~
c -d a ~
a~
v~
~

o ~ ~ Q ~ ~
~
~
~
;~
~
p 2s 'b a, ~
~
fl z U U c o ~, ~, ~., ~ tn C ~ N o ~ -C oa a.
~, ft) Ca O~
~
N
i.
~
~"
M
f ' ~
~
~
' . -. ' r"~ ~.
. -a ~ O
~' ~

+. . ~
p >
*' M _~
. . 'D
' N
'~ ~n '~ N +, p .O
O
C
.~
b ~
x ~
.
~

+~ N ~ ~
o_ b "~ .~
~
~
~
>
~

+U-., N U ~ y~-, ,.,., of ~ ~
cCi .w ~
~
U
~
U
O
'U
~

''~'-' ~ ~ ~
~ o ~
U ~ o U
~
~
~
x ~
o . ~
~l ~s~-o E~~~ .
d 3~L~?~Qda.
~
~Q
>A;

0 0 ~ ~
~o ~ ~; ... o ~
o ~
'-' o j, ~ tn ~ ~ O ~ O V ~ .4: O ~
ti7 ~ ~ N
v~
~
v7 O
O
.~
,~
O
.

~
~o~~

b H i ~ ~ ~ ~' ~ ~ 0.
' c~ U
' ti0 ~
"
~
~n >
a~
o ~
'"
' ~ ~E~ -~~b i ~~-a ~~ a :b.c ~ > a, :~,~
~;' i~
~ ~ _ ~ ~
EH
~~
c W

.~,~,~ i,0. ~ ~ ?~~~
~ ~ .flx ~,~q o wo a~ E ~a .~ ~ ~o, .;
. a~ ~... o ~, ~s ~ ~ . ~
,~
~~x a~
, wo a~
~
~
~
.
a~
+~
, p b .
c,.., .c i ~ o ~ ~ ~ c~ ~ ~ b w.
n y w "~ ' '~
~ ~
~
"
' ~
y ~
~
.
r' ~
a ~
f~
~

~ ~ , o ~ O ~ E-~ ~1 . o o U O
'~ ~ ~
. b~A
O O ~ r~ ~ a~ ai ~
c_~ O
~ ay ~
[-., Gq ~
~
~

O
~
U
N

' , s~
~~"OO O ~
0 DG~~ ~~'i00 ~ ~ ~F".~
OR 'O
~~ Op ~
O
~

~ ai ~
. . ,~
~ N , . c .
.C
O

O .~ l~ N U ~ ~ ~ Ga ~ G' N ~
~ W O ~O O ~ ~
CU ' ~
~ O
~
l~
N
~
~
~
.C
~
N
W
O
~
tiD
' ' U ' O O t.. i a N N_ ~
" ,~ +' U
w~~~s..~ ~ ~
UN .~U.U'O *
~ 0 i U~U~ ~
a, U
~
N
~a'' 'NN
~v7~W~~U-~~
-UN
~
' ..~ .f F~
r a ._ ~O 4 4 ~ ~ L
' 1 +~ c~
c~ ~y~~O
~ '~O
~ w ~ ~~
+..
af ~. ~ ~
~..f.' ~ ~y ' .>s.. ~
~ .~~s. , . ~
+ ~
O ~ y., ~ * U
U U O b ~ '"' N
~ "~
~
~
~
U
~
O
~
U

G sw_n s~
-~~.~n3~ ~ O
=~o ~.~p cC
v U-~~~,o~.~ C
~ 3 ~O ~
U ~ b4 c~ -~a~.~3~' f'-' 4 ~o~.
cct H ~ L1 v f3 mcS
'~
E"~
c1 U
O
W
H
~
O

~ ~ ., .
. ~
it . .-~ a.
c Y.
.
..
, c c1 O ~ O ~ cn O

c~1 . M
' \OMO~ lpM
O , . O
O O ~ ~O M .--~
O
Op ~
~

333~~ ' 333a~

~n o , O
as Q z~

d a ~
:.
w c~

.n '~~"
o a .~
.a .~
cG
~
U

~
~
O
y ~

c ~ o ~V ~U
O a _ .~.
_ W
U ~.., N Ga "O a1 ~"

r'U~ ~q~
c ~
c E

o. ~
o ~

c c E~
~Na _ o~~.
G~
-o~U.-.-o~W
~

U .
C O
~ ~
O
.C
U

N .-.
~ ~, O ~
a p O
E
a .+, '.O . ' t b0 ~
~

N
~
~
~a~o o0 wQa ~~ ~ raQa ~p~~~

o c ~ b u vi U O

.
U N ~. Q Y ~, ~ "'~_ U O r.U. ,.C ~ C/~ b..4 ..~. " .
..C . ~ .~ Q
Y
~ c ~
~ . ~

r--~
~

.g ~
d~~ >, c ~ o ~
o o . u. .
~ ' z ~ ~' ~

U ~ z ~ U ..
' ' a ~
U .
U at ~ GC N ~ .~ Y ~
~

L y~
. s. ~ ~ , . . ~
N
~

O ~ ~
~ O :~
O ~ N .N ~ N
~
~
w ~

U A, U W
r a as c~ a x x ~ x V ~ i 'C M (Y7 V7 b ~ ~ N a1 l~
~

U N t-. O ' ~ Y ,.~ c~ ~' ~ v ~ s.. ~ z d~ o ~Q~ ~ ~,~~~
WM

.
' ~
U w a;
~w~

z~ oy~~ ~ ~ .
Y ~
~~a ~

o.r 'o '....' .
~aj .
~ ~ ~c ~ ~ o o~

~'" ~ O ~ ~n ~ O ~ V ~ ~ ~ O~
~ . , Uj ~.r U ~ ~ ~ .Q U U i .
a '~ an a~ > N U O W ~
' ~ ~ w ~4 c ~

~ '~ .~ v~
~ ~
~

2s~U ~U ~~ ~~ ~
'~d o ~
~

>, :
~'n ~- oQ~ Q , ~ o,~~ o ~ a ~o ,.s~.~a cw~ o,.~~ '~
~o ~~~~'o~x~~

Y ~ '~ 1.~ .~ '-4 ~ ~ ~ ' ..
te .pr ~ .
_ ~ a .
U
U
p x ..~W .~?,~ c AW ~ ~0. r l~G cE-n~
~

Y
o ~ b _ o ~.a~o~ 4. ~ ~ c ' 'O U +u ' O U .~.
~ N N
N . ' ~
~

+r Y
bA b i: ti b ~ ~ ~n ~ U
y b0 vi ' V O C.
fl ~ N ~ '~ ~ U , ~

. ~.

~ ~
~
v n ' ~V' ~ a,'~'~
' ,~' w o ._ ~ ~
~ o ~
a~

n ~' ~':o o: n.c a'o ~~ ~_~ ~~.~
o ~

. ' ~~~ +.~ Q U
~~cQG 7 ~ VUcUncYCQ
Q ~~~~
~

. N .
cct ~
c Ur-.
C ~~~~~Cd~~O~~
UC~CS'.,'k' N O V ~ ~ v .~ U 7 '~~' !~.' V
.~ ~ o ~

~0~'~'~~ ~n.C~~.,~,.~~~~~~
N
~

c U S U .~'..7 U ~ .4:
a '~.:' '~ U c~
U U U aS U O
~ U ~ U

N

a1 ~ N

o .
? '~
V

~O G ,~
~
c~
d o ' o~ o ~n a~
o Y
~
a a o ~ ~
U
~
7, ~' ~'~
~

w o ~ ~~
. c~ rte' v x ~ ~ a _ ~ 7 ai ' .fl q C7 ~ ~ or.
c~

N -j U
' N
" O .C ~

y ' Q O
U SON
cYCU

F
,, .~ ~ ~ U ~

.
o~~ "' . _" ~
~
:
.

...~ _ oU ~
,~
~
;

'-, .
wo o : Y C7d~
ff o Uv~
CC~Un .V'~ ~
~

~ N -p pOp , O (~ , b ."'~ V1 ~

cG ~ O O . ~ ~: O . ~. O > .

~ ~W ~ ~
Q

U E,. U~~a , _ U ~

o ~ ,.

p UU

w ' .
o o WrJ a w ?: U o + a ~
4' o~

~ 0 cc ,.c~ C
: ~, Hi '~; i~. o ,~ '~t '~ ~ ~ a~ y y U +.'> ~ ~ a . o~b~,~ a,o=a~ . ~~~,~~.
E U
f~ -o ~ E-~ i ~

' ~, ~
~ ~ C, > ~ c a a~ a .~ ~, ~ 0.1 ~ -~ o ~ ~ Z .
. .~ A
~

~
.o , o ~ w ~ ' ~
o ~ > ~ ~
.x ~

C .
~ ~ 0.1 P, ~,. 'u:. N '~" G' ' ~ ~
a k ~, at Gw cV
a3 z n. ~ ~ .~ ~ O
N r Vl ~ ~ b ~ ~ ~ ~ ~
i"' . '~' U U

~

. ~..
W ~
~L~
W~
' ~ U
~ ~
~ p p., ~
ca , . n a;~
t .
i ~ o ar -o ~~
o on ~
~ 6 (~.Ud;~.~O , ~, v ~..f~.wnG'~c~MOGS.U~cC

cG t-, cC at ~"~ ~ ' V7 c 'O ~ U
~ ' 'a' ~ N ~ WO
M U

. c , _ +.. ~ .O
E-"c~c a.~~O'~N
~~~~ ~~N
NN
~

. O
:n ~ U o ~ c ~ U c i Q
~ -~ ~ f ~ ~ ~ ~
~

" o ~.
U
w ,~ ~ - ~ d o ~ ~

~

~. ~.'.' ~,>>., C1. "' ~-" U ~ ~ p y ~

~.b ~4~ ~.~N w ~ ~~23~ y N

~~~b~~~Q~, NY~.~x~~

pU.~ ?~~v ~' ~=
~G~~ ~
~

c ~
,s ~
_ G
~. o a ~ p cC
~ O w1 '~ ~

., in ~ ' V r. .C N
O .~ N
G4 ~
C~1'~
TJ
~

O ~~U
. ~.
O .C>, ~~ p of O -~ O j ~"
'~~' ~. U V ' U

.... ~ "
~, U ~ . d a .
~ ~ O
G ~' ~

~ .,~ a~
n .
U
n o c~
d d a~ c~
~ ~ s o ~ 'x . fl ~

a~ ~
N .
n~ ~ ~
U o=o ~;
~U :d ~ c ~

. .
. ...
.

o , U ~~
.~ N

r, .
N

O

U~

yr N
N~

i .n U

.--. ~, .

UQQ

. _ ~ U ~
v ~ U c U , c .~ y.'~
W~UO
o a :~ ~ N ~ :~ a.0~ CJ~ O O . V ~ ~ U ~ ~ .b ~ O s. O
_ U
~U ~.o..fl ~~ ~x.~ o.+~ ~ ~ ~n~
a~ ?, oo ~ v~ n. r; tr ~ ~ ,~ " ~ O ,~ ~ ''J .x C Q as ~ ~ >,M °Q o oU~~~ o~~ ~ ~..xc~q .o~,y o o ~~ ~ ~ ~~~U ~ ~d 0 0 ~ ~sv, x ~ ~ ~ ~ ~ a~ ~~ ~d o~ ~ ~..o~
..-. O~sU..~G OQpp~.~.UUS.O,~O~,~U~N
>UQ.n n~-.',=, > ~ o.GC7o~ ~U ° ~.c~
~U,~dU i?OWdGs s°'. ~ ~ ~ >v~.-;°~dd i. v»n N U U
a~ a ~ v U d G~- ~ .
y N p ~ a..u U .fl ~ U O ~, w ~d N Vii'., ~~ , ~ O i~ C v.~ ~~
° ~. ~' nn ~~ d ~ .~
>:'.
Q .~ 'd ..U. v~
y V v7 3 U
7.~~OTJ~T30W~i0 >r ~ ~N" .~ '~ J,'° ~ c~ y b V bD ~ !~ ~ O ~ c~C
4. ~ .p U
~ a~ >~ .~a U ~ ~,~.~ ~~ ~ ~ v ~
d~ o ni Q~
N o0 0o yn O O~ O
Olden- OI
x az.
U N U ~ O, a U U (C V'1 l~ ~7 _O L~' . ., cci p +..
'~ U c~G .'O :d ..i"~'y., N N ~" C ~ cQC '~ O ~ O ~ ~ ~ ~:,~
U ~ add U;~ ~_.~ ~
'.~'' O N U" ~ O ~ '~" ~ O p ..fl l~
p. ~y: O_ c~ U o o ~' G7 . .
:~ 0.~.~ > ~ ~,.od >,o> O~~d Ud,~~ > ~ °'dd > ~~z ~n~°d U
.L
U O cC
r,U. O
U ~ c ~
~ ~ ~ ~ H

UU..°aUUU
>, ~ +. .. o ~
_°? U ono ~ o ~ ~ ,-.
°' b o~ a.
° ~ i -~ o ov 'o .~ o ~ Q ~= ~°U~ ~ 'o t~j o ° ~ ~ ~ ~ ~ ~~ ~ ~ ~ N ~ ~
~.o ~> ~z~~~ ~b U cV N cct ~ .i-. of P, ccS -.. fn 4~ ~ N
~U O ~ cd~ ~~~-'z ~ o~Ob G~M.~J~'~OO
y C N ,G ty ~ ~ ~ ~ T3 ~ o--a N ° ~~ O
°,O N~~ ~ .D O U~p.., ~N ~y .~.,wo +~
~.nOaa, ~ .iW~ o on~U~.o'.° ~ c~ ~~..;,n -~ E... U yo ~ p ~ .~ 'n ~ ~ c~ ~''~ ..~~. .~ TJ ~ o ~.c~Gt.~,~~~~'"~y~,~~~~.pc~cv p.., :p ° s:. ~ al ~ ,n d '~n +~ tp ~ Pa o .~ "c .
o ,~ ~' > -c cn ~ o o ° ° y ~ .r A-' a~
'.:n ~ U o ~ Q U c~ ~ U -a ~ ~ w ~ ~ v o P, v~
~ , tn d c, "-, W o ~ ~ ~ 4" W a~
c o c ~o '~ o U ~ ~ o U
° .~. ~ >, .+, U ~ ;a U
p°~p ~ v ~ ~ ~ :b ~ .~ ~ d .~ ~ ~ ° o ~_ ~ o ~n .C '~ +.. v aWG ~ L_~ W +~ .~
~, ~ c .~ '° .c W b ~ on ~ .~ $ ~ ~ '°°
w ca~~~'a'~°'at,~~,.CaW.x°~ Ua~
~U 3 N ~~ .p O pl~.~ ° ~~", d ~ 'p ' ~ ~ U
° ° ~ ~ ~ .~, p W U ~ o U . v~ G C r~ ~ v c~ w U ~ ca ~ d a~
M
N
_N
N d ~ ~.
~ ~W bO
W O y:
ccj."~,~° U ~,W 'F~." O'~' ~ W ~ ~" ~" ~ U CL
o~p ~~-~ ~~ ~,~U ° $'C UH ai °U
~ ~ '~ -o ~~ .n o .~ -~ ~~ .b ~ Q U
o . U °
W ~n ~ ~ °.o ~c°o ~a~.°o~'~~'~U UO
.~ OdOU+~~.OC~CU(~C~G~c~GO~~1' y°.,~' on N
.W n ~ o ~ ~ ~ ~ i cct c0 fn ~n ~'d~ o ø''~ ° ~d~ ~'~ c ~'~E" °'r.4 0 o W ~ ~ ~° ~ o ° W -o ° ° ° a a~ -o o U
.U,-.;oU c~ t~ op oU~ o " o ~.n~ ~naU
~W°'~~W~or'o~~~o'ro°'~;,=, Vo~o U~~doa',Ud~~ ~d ~ ~.~W. ~.~o°, coe' UQd ~_s . ~s : o . U
' O
't7 ~ i O ~
O ~
O
O

. .
O ..
U .
U s i-U ~ ~ ~ ~ ~
U Ir ' ~ ' ~ C
>

._. , .
,~ ..
O .~
~ ~.:~
, ..
i. Z ~ ~ x c~ ~ CJ~

_ ~ ap ~ ~"
vpU~U
~~"~'P~ V

,W "
n TJ ~, 'y~ '~'"., . ,~, U c/] ~ CG
U
+~
.r..
~
i-.
' ~~~.'~~o~~'~~
~~~a _ 4j ~ O Q F.y p Y
p" cG ~ ~ O
a3 U ' G." O
N
?
b ~
N
+..
O-t1, t1 fn ~n tn O~V~~.~V~~i.~.i.~..0~. ~~T~~U,~, .~.,~~p~~cC

a, cn r.~ cra > H c~
E-- a~ d Ca O ~ E~ x d La ::
O
w E-E-~
a7 H

i O, ~

+.
U
O

y ~
Q Y
~
~

.L"cct.
U .t O~,'~N~ ,_NN
U ~NNU~bd p ~
.O
O
~
O
~"

~ c C

~
.~.'~ x ~ .c x .-, E
v~

~ ~ ~ +~ ~~ ~,' U
CcJ M
~ .
~ ~ ~
~
i:
~
~
O~
at ( ~ O
Y ~ p O
, ~ d ~
~

s.. ,ti c~
~ p >
~ ~
, .,r . G.' N ~ .
'.~,, O
~
y E-~
' TJ~~~ O~N~'Opm .,~C
c~

0 ~ '~ 'c p~p ~"'~
G ~ w ~ N
~
'~
~
~
~
~
~

o ~ -a r. ~' R ,~
A ~ .n ~
~
.
~
U
~
' ~

~~ ,W.?~
~
c.
~
~,ou~
?

~ O U~:.
Nd ~

. a .o ~
~ r.
.~ ~ >,~ ~
a: > v~
v N

, x cC y ran O ~ ~ ~ .,_, O ~
p ~
~
Q
O~
' ' ~

.~ Z ~ ~~,~ ~a ~ d ~
~~c~aC7 o o ~

p c~
Q O .
.-w O
N

N
~ ..p p Q
O.
~
U
L
~
~

p 'O b e~ cC ~., 4~
~
~
"!-.' U
c~
...
N
c~
W

~
O
~
~
V
'O

...
.
,i,~
O
Q
U
~
., Q
Y

..O
O
~
,fl C
m +:, ~ Y
O

~
Q .Y
U p ~
U
t3.

~ :,O ~
4a ~."
N
~, ~

-b>' w.~~
~-' ' .
~
~Y

4~
'~.' U
ai Q

~ Y
p ~
Y
C
"~

+ .~.', N dw~
~
O
c C', s.
.' ~,..' d~~s~...~~3 N

p O

d.

M
.
N
o0 r~

U P
C a _ _ _ _ N

o o cn cn ~
t~
~
~

x m ' N .
p ~
~
:.
T
~
O
~
ccS
~
a ~~
~M
o _ ~
.
y...
~~,.
__o Aa O

d cd Y
~~~
~
~
aQ~~
~~
~
U
~
H
~
o o >, d ~
o ,....
p ~, >, o .
~
o ~' -~
~

o o w o ~~~do ~~:.~
d ~

:
rr~Yo.":
U' N~
Yo _ ow ~g ~
o ~N
~
E'~~~
~
~H
~
EZ

..o ~r' ~~
d ~00 o ~p a ~ O.~j.b ab cL~ Tp , ~
U
p oU
p oU
o at~
o .
~
.
~
O
.~
,~
~
.

0, ~ p U
~ ~
O UQ
~
O
~
Y
~
C

O
(~
A
Q
UQdo~Ud~~d~~~~d~~d~

.

o O
.
U
S..' r.~, G".
~~
Q

to O
~
Cl.
~

.
~

_~
O
c C
O

Vl ~
~
V

C
~~'~'~.0~

4r .~~ ~ ~ cG
~ U
~, ~Q
~b~~

~~~ b oa~ ~
~~

~
~.
~' ouY' ~
Gay, '~ N ~ O
~ M ~
~
~
~' O

~ ~
~ ~
~
~
~
~
, .

O Y
~ . ~
. U
i '~
, ~ o v o 5.
o ., O U ~ ~ ~ ~
-.
~L
cct ~, O
~
~
c~C

~ ~ U o ~ ~ ~
~

o ~
'~
~
~

" ~
d~ U N .

( .a;
] U U1 V
~
~
coo O
-~O

N y-i Cue." '~ ~

~
~N
~
H
,.-n ~

4~ Uf~~ cC ~7~r~
N'O~
~O

(.L~] N~ OG~~~N~~

~
U

N sr' wn .;'"" V7 O ft U
~
G~
U
.
.F-"

~: ~ ~x o ~
~
o w a ~~oN r~~~~
~
~zN

~

'.'.' ~ ~ O .~' .C w .
U o7 O
O

' ~ o , -'a ~' ~' y" ~
~' x ~y ~
b ~
~

a..' O
~ :~ U ~
y ~ -tFn~ C~ Gi O o-.'~
' ~
- ~
Nbt-.i.UN47~y.0'NT m''~
NHO'TJO
~

.-.p0+> >7v~~+~s.'.~ .
U m~
~ O O
' ~ p cUn ~ p ~ 'O' ~
Y =
0 z..

.. t. r i. U
.. Q ~
t~ t,. U
. s O
~
., ~
~

~1.. N ~
c~C O. ~ ~
~' ..~. ~
' pp ~ ~
_N
~
~

a~ ~ ~ d .~
~ b4'C
~,0 O ~ ~ ~
' 'C
a~
~
ca ~

~ ~
'* V
'n~ p ~
~~
O

' ~O
rN , ~, ~
-~.4:
+
.~
~
o ~' ~
.
~

~
~
a.~
c ~
n ~

~, ~ o ~ U
a~ ~ ~
~ ~. O ~ ~ b ' . ~
7. ~
~ i~
~ a ~ p., 2s .d ~
' U
P

. -n r t... N
C7 ..-r U v~
O U C ~ N ~ U
c~ ~ ~ C~ ~ ~ ~ y U ~'. CJ
c~ ~ ~~ ~
~
~
O
.C
O

~ .O ~ U
~ ~
U~ N N
O cG
~ ~
~~
O
~' ~
~
~

~ , U ~.
b ~
G _O ~' c~ x V ' ~ O O ~ d N
p ~p ~
~U f~
0 ~
ON ii "O O
~ ' G
W
w ~

..
' t.
.~ , .. , .. ..r + , + ,-. U
.
+

N
a1 N

O . ~O
N N
O
cn ~
D, a>
a\
a1 N c~1 .
c0 c~1 o cW O

o o n , O O ~ oo ~

~N

zxx due, U
U
~
U

.

~lU ~.~,LIQPa U ~ ~
~
U
~
U

"

~ O
~
a O

b v O q b _ ~
~
+.
.
.
A
~~
~
G
~

UQ ~ U
E~~dC7 ~~

_ U ~

as o > ~
nn U

~
1 'O oo ~ ~
W O\
fl Y
O

~ ,.
cC 4.. ~
' ' ,..., s~, o x '~. ~a a~ a~
. ~ U .~ ~ o n. 0 0 W

o;o~a~~=~
o~.W ~
~ o O'H :o 2y N
,~.~~ ~~.~a~ ~ o~~ ~z~ k~~~

ca o bn a~ ~ .~ ~' c~ ~ ~' u.. U c 'a N ' ~'~
~ ~

c~ ~ f1, c V p ~ ~ ~ ~ ~ 'X W ~ ~
y~.~ ~ ~p z ~~~
~

~~ MO .~ , O ~ cd .y.--i'oo ~--, aS ~. "Cf P~ +.
.~
'~
N O

~ v y ~ "
fl >
~

..x p ~, .
~ . ~ ~ p,, " N ~ ,.
C ~ Ls. O
E''~~ ~., ~
~
~
U~

~ ~ a~
~ ..
~
o ~~
~v, ' ti a ~, O V G m S~ ~ ...U.
E-" TI ~ O
~ ~
~
' ~n ~' ~.~
x ~' ~ ~ x ~ ~

a,b a~ ~ ~~ a.~ ~c o ~N'~~
~~n o ~p;,.~

O ~ "'..' > , ~.~' ..~ ~'~ ~~ p O SAC ~ ~
fl ~ >, a~ a~

u: .~vb a~~w~ ~ vo.v~
....,U o aUN

.~ ~ .~ ~.o , ,~ 3 ~ ~ :~ . .
~ , s.'v~-' ccn ~

~
a x O ~ U
' O O O

CJ
O ~ N ~,' +~.., ~ CJ ~

~ a, ,~ ~ >, ~ A

, ~.

~
a ~ ~ o-~' ~
~

U a ~ +~.. ' N~. O O.

O

cn N

.

O
tt O

r. pp O N ~

O O N

I
n p., P-~O
Z~~, N

O ~
.-. N

... ~ , U .fl o ~ ., N

r, , ~n ;~ U . , r1 ~ 1 , WO 01/79442 ~ PCT/USO1/11850 c ~

o E~

~~
~
.~
o ~a ...

~

U ~
U
L1. T 0 ~ .~

l1, ~ C%~ H ~

ran ~ . ~ N

c~ ' ~ ~ ~ ~O ,.~ '~
~
~n+-~'~3a~

~ ~ ~W~.~ ~ 'q N
>
Q

, .
'~ ~ .=
~ ~

, ~O .~ U ~ ~ ~ ~ '~ .'~
U ~ ~ M

~ O N . .S''~..!' O U, .~
~ .

.
~
~

~ bA ~ y, U ~ C/~ ~ .

U U C U L' ,~ ~ ~ ~ "~
~ O
'O, O c~ ' r~

-d ~
O~

U
O
y." c~
~ U ~ i~ ~

.

V O ~ ~ ~ O

~ ~ ,~ ~ - ~ '-' ~a~~~~~~,,.~o'bo ~ ~ ~r~xz~~ ~~x '.
.~

a-o ~ . O ~ ~ >

O ._~ ~ O Q ~ O c~ ~~,~,.. ,O ~ v~ ~ U
a~

~ ~
~
~

a~~ N
c v~~U nn c ~
aLO~
~

C .y ~' O Q .fl +~ .F ~i-1 ~ ~ ~' ~ ~ ~ ~ ~ ,~
~ ~ '~ ~ ~ ~ ~
~

-' ~ ~oc~o,.~U
~~~~'; ~~oa,~v ~.~~a cc~~~c .
, ~

~

,.C . .~ ~ >, c~V . .o at _ dp .,..
~ ~ ~ U .... 00 ~ ,r ~~
~n ON
~~~
N

~
~
' ~

~~.- CJ~
~
~tnO ~
~
~.~.
~
~~c C~~~~
.'~~
'~
d ~

,.O Q7 L: 0 l.
~ ~ ,O .C ~O .
C ~ N ~ U ~ U
T c UU
U
~
~
~
~

"r ._..
~~vy.~-~ n~y ~
,Q, ~~
.,~ ~,,.,,CDy. O~
U.~~
O~U~.~
U

CL~.. ~OOG1 ~a7~y~~~~.~~~U .~;yU.,~TJ~ OU~USJ'.O
~ U o~
~

, , _ O 'C W
U ..~ ~ O ~ ~ a O ~ ~ ~
~ ~ V ..Ø ~ ~ N
N ~
'~ ~
~ O

N ~ .~
.. , . O
.C F
ro ~
~
~
U
~
U
~

~r. p.,~'~~~N~
f~r' '~,~.' O
r~V
~~OpO
00 Y~~
O~~~~~

n'~j '~ o > ~ ~ > ~ a'~ _ Ll~~~t ~~o a '~ 'c~ ~ ~' ~ o a.~~
~ n ~~
~

~ " ~
,. ~ O
. U ~
~ U ~ U ~ ~ x N N
C U
U
~ U
~' N
' ~ U
U ~ U U
" ~U
~
U
'O

. -S U O
.. , t , .n 1. U
., 7. .
. .r ~ ..
~ .
. S
S i .

N

~n N ~D cn N , .
O O cn n1 o0 cn a\
O ~ N ~ N cn d' ~w0 ~

I
I ~ I~ ~ ~ ~ ~ ~
G. Ch ~ p. O O O O O
O

zxzx~.~.~~.~~ .

y, cn o ~

a UQ ~~

O ~ N

a_o..oU~
. , , _ _ U d ~ d "~

WO 01/79442 . PCT/USO1/11850 -c t ~ o o r. ~ o ~ °.
cC T3 .~ '.ir itS i.: U' ~ ~~ ~ it t' o a~ . a o W
'~" ~ O "..~' '~ ~ > '~ ua ~ , ., o °~' z ~ .c c°a Q >=
~r~ ~. ~ ~o~p.' oU~o ~ ~.a ~ ~ ~U."U-o.o o ~ a . " .~ +~ +~
°xQ m L ° ~ L '° ~ ~Z °°aT
H.° o ~ ~' .r~. ~ ° ~ o ~c~ ~ ~ ~ o Qx>~E-.~~>xQ~~...,a;HUU,..., axxoc~xa~aar~~
a~ ' o ° .~ c~ Q >, o w o o.
'o ,~ p o 3 nJ
O
v ~.
O ~ G~ . p ~ ~ ø~ ~ W.~.
~ O ~ O
at a ~, C ,~ ~ '~, Q ... G-m,~
U v~ 4'~-. ~s ~ ~ .
° .o o ,Wo ~ f~ v ~, ,-.
~.
~ ~ ~.o.~V' °-~
c '~, ~ o a~
ar~~ ~-~,~ ~o ~"~ ~a~
o~b_n.Y°'.N~~ '~~3(~
~" .~ ~~" U ' p .U 00 _ U .b~DO C O , W ~ ~ v~ ~ ° Z N f~ ~ U v ~l~
E-~ a~
~~ ~
o ~ pa frl o y m ~ s.~.
~.o.~ ~ enN.~ ~
,o °~' ° ~ U o +~
. .~ ° ? ~ ~ o cC ccf s-U. O U ..fl .O ~ a: bD > ~ .~ f3.
y C ~. Q. ~. f-' ~, v~ . ~O ~ ~ v~ ~
O ~ 4--~ O U N 00 '.O N ~ N O ''r ~ ~ c~
.~,UOc3 UU~~U aSb~DC~J~ 4"'O .~.
N v~ N .O ~ ~ U E"'i O ~ ~ .~ ~ N
',r>,c~~>,"° ~wU~~o~
~',~'a'~o~°~MO~ U~ c.~°~ i b a~ a~ ~O w U ~ r! bn H~'S~.U;~c~a~..ac~Gc~ Q~UU.
M_ N M lp o0 ~
,_, ~-. O~ M N O~ O
O ~ O1 .O I~ ~ 00 O ~'..' ~' O vD O~ O
O Or'c'~Od'N
C~l ' ' M .--n 00 V'1 CO
M ~O ~ V7 U1 3 W~~~~3 ~ ' , c-N G1 ~h N ~ ~p ,n U N
,'~ N ~ o o .
U z~~~
. . ici T .+ . N s~ :n N . .~ -o cy o ~ ~ a ~ ~ .a "~ ~o ~ ° ,~°
a'.fl a~~i.~ °~'~v~° ~ o . Gd ~w~~'~~,~
y ~ o ~ o U W ~o ~ U ~ ~ .~ ~ i ~
~ E.. N ~ ~ '+; ~ o ~, a~ > p '~, ,~ 0 0.~ ~ s~.._ ~ p"
c~ as ~ -o i~ ~ ~ o ° >, G C-.~ .» ø. W N :~ ° ~ ~ 'b '~ ~ ~ ~s x UONd'O~~"C7~ ~O~ W~~~~ OUpO
U~.~~a,cn N ..,0 ~NN~U7~ ~ a ~pa~; .oG~ ~ Uw ~'~
Q G '''' ~ ~ O ~n ~+~ p ~ i +-. 'C7 ~ ~ U cn N ~.~ +r ~~; O ~
~~d TWO o~~ ~ oUN ~ ~ ~ o~ ~~U~ ~ ~ o 0 N ~ iC p..~ N ~ '~ O cC O ~ c~ . ~ ~ ~ O ~ ~ iC 0 O C ~--' (~ N p C C cC ~ c~ ~ N ~ W ~ ~ N U V
v~ oE-U ~s >,o o ..,>.oH ~.~ ~1.~ 0~~,4~N k o . - Ll . ., ~o -~ ,~ -d .-. ~ o ~ ,-~ .° ,...
.o U ~ o ~''~ ~ U ~r ° o ~' . o 0 o O o -° c> ~"' ,X ~,~
° o U U ~ U s Q
N ~ '.~. C Q Q ~ ' '~ ~ ~ N N :~ G ~ a N ~ ~ ~.,_, ,+'O~".~ ~ M :.O ~ '+.~ t.1 ~~ .:. 'C3 >
~U~a.~ ~~,.wa ~o ~ ~~~ ~ ~HQ~UQ~ 3~~~ ~ ~d~ca ~~N~

~ E ~

z a .~

~.

~ ap x a, ~
o .:o nn~~
O
~, a.
W ~
., .

U ~~
~ i"~
, C
~
N ~ ~
b I~

~
U ~ N
~ ~, O W
~D

N U pn ~~' .,~
M
Q. v ~ O
~ ~
~D

at T~

O ~ , V .O
w ?~
U ~
,..
~

~ ~ ._~
~ ~
~ Q
~

.-.

y ~ C L1 ~ ~
N c~

~ o .o U ~ ~
~E-i~~~

.

o ~

~"' U
~

S.. ., .~
..
~

~" W
a.
y ~
.+~'.
i~-.
0.4 C

r .
O r~'' i.' w 3 ~s .-..
b U
U O
+J
'~i ~ O

.C
U

U ~

y ., , w O ~ ~
.~
U
p .
N p p +.~ O
'c3 ~ C.
'+.~
,-.

U c~a v ~?'~
~ v M

N

a, cn d, H

~ O ~
N

F-, C~
N bT

. i--I
'-' +~ W
~
~

O
N W

N a .., ~
U

_ O ~ ~M
O x ..M-n N -O
U
. ~=.
~ F~' n-, ~
N p .+=' N ~
~ N
~QQ ~a~
a ~

23.

WO 01/79442 ' PCT/USO1/11850 C G U O C , i.: O O ~ +.. .t_'. ~ ,O
c°ac~a~"~u~ ~ ~~ .~aCj a~~o '~° °~-°~ Q,~~ c ~ o '> o~'i °' ~z > ~'i o ~' on0 . ~H o ~: z ;~'+j ~s~U ~ ~'O aw' lj ~, ~ ~ cn ~ U .~' - N ~ . " ° +~
_~~_~~~~.~~~~~x~~x~~~~ ' +' v, ~ .r, ~ O ..~ ca ~ y '~'' O ~p ~ a. m ~ ~ C O
... U N 7.. N N .... f..~ 7.., ,~ N O ]m~ CC cd ,...., ° c~ 7... ... a5 ar~ax~,xxr~xHa~xa~c~UHZUC~r~UH
N -a , nn.o..nO
~ o " , . .
o ~. ~ .T c -v ~ .
~ ° H o W ~' , ' >, ~ ~ y .
o~>,,i ~t o c ~ ~
~ ° a~ , ~.
a ~ ~' ~NGa ~~ a~ _ , o Ca x rte; q a., . , o~ a0.1 ~ ~ ~' .~ ~ E-, ~ U v .~
a~ °
.,'~~c~~ T~~.?
U
a~Uvax~~,'~
o b ~ ~ ~ ~ ~
U
v,~ ~ orx z.. a~ ~ a~
b~A H ° ~ ~ .C
~° .C ~ p ~ '~ ~
+~
cH ~ c0, '~ O N~ ~ .
O ~.~ ~ ,~ ~ar ~.' al:-. , ' a~ 7 ~b4 pC, , a,~""' 3'U °~'U X . ' ~ ~s ~o E-Ga:cU~~ o~ ~ ~
U 3 ~ ~ E-~ 3 °. °v oa m c- N
N °~ oo ~n cn ~_n N oo a1 c~ .
N M O N
~ ~ ~O v0 N
a1 ~
~~333 cn d' Wit- ~n va ,-wr ~D
N N ~D N ~ vWn CO
I~ l~ ~O t d' dwf' oo ' , O O O O ~O v0 v0 ~-~ , O O O O O O O O
OIOIOIOIOiOIOIOI .
~zzzxxxx U ~ ,~ . " ~ ' m .~ i CU
" ' C ~ " p ~ ,-, , ~' ~' N p c~ ~ ~ c~ ~ ,~ a ~ V ~ p .~'~., Q ~"' O ~ O
U ~ N (~ N O N crt ~ p ~ Ec..~., ~~ U iY- .f'~' ..I"'~ w ø, vp '~ c~C ~.' .'fl ~ ~ O
~N ~U ~ a.~~ rxnx ~~p '~° oo~tx0 ~o~~.~~'~ °W~q~ coach ~'a ~ O ~
~ ~ '~. ,.o o ~ ~ ~ o N c~ an ~ d ~ ~ ' . A o er M '~° or, °~ o o i ~p a o ~ ~~°,°~~, ~~
o~'''~.~ o oT°U ~,..~.7 °O o °
i> ~ : D U
oQ0 ~ ~U~ ~ ~ ~,~ o~ c ~A.~ ~ ~~M~ ~~~~ ~ a ~a?~EM- c°+, ~ ~U
' > ~ ~ c ~ ~ o o ~ ° ~ ~ ~.°u~~n ~a ~U ~a c~ o~ °~
° o ~e o , ~~...,~~ce~~c3o~~~~C.nao~~oi-:V~~°~'(L,.,~,ca oq o °~ >;>,~ o oc~~ ~~ o~Q ~,~~ ~.~ >,o ~~Q ~ 0,4~~~..W~ >;e~,~ o~Q
U o ~ ~ -o ~o U o U o~ U v U c~; o ~ H ~ ~ o o '~ U ~ o o ~ ~'~. U o v U ~ U
~i Q ~ a ~L ~,~:9~~ o o~~ ~~G o~a~'~°'~ ~~~'~ o ~~~'~~ o o Q"~'~'~
~~'~~ o'~H
Uda~UGO ~ ~Q ~~a °;~'aWaO c~c°Ooo~ ca ~ ~a~ v ~ ~PaO~ ~~a ~a~~

L
U
U
N
,.. ~ U ~ ' ~ .~ >, ~1 . od>,~ ~o >, , O ~ ~ ° .~ .~ .~
U
U N ~ N
O ~ U ~ ~ ~ ~ ~ ~, xU dawaaa.~

.~ L ~ ~ ~ _. ~ ~ ~ oo ~ p R ~ ~ .~
°' ~ ~ c~ ~, .5 ' v .~ N '° ° ~ O ° ~ c~°
o~°o U y , U 4-n ~ ~., O ~t ° p ~ N i .~ o ~ + en ~ L ~. o ° '~ ~a U d' ~ o 'o ~ ~ ~ o ~
o.~ d ~ ~~~,~,a3yr ~ ~ od ~U ~~ , ~ ~ M v1 _ a_-.
a~ o ~ ° o ~ v~
p'N VUC~~~NCns. N
T.C dD O U ~ OV ~ 3 0 Sy U O ~. L ~ ~ T ~ ~ ~ ~ ~ .~ ~

a~ L ~ ~ ~ ~ ~ ~ ''' on x ~a ~,, a ~~ ~ o~V~~v ~~ ~~~ a.~ 3~~7'cc~n~ °
~ ~ as ~, U o E o ° '~ o, ,u. '° -~ r' cv o .~
o,~ ~o ~ o~~ > ~ .., o N ~.~ a0 o M o ~ o +~ > a, ~ o .~ ..c "~ ~ . ~ ° ~ ~ ~s ~ ~
~s ~~ °'°" °°~ ~m-o '' O~.ox ~ ~~f °-o o~ o c~~> ~.~~ ow~G~~~r~.d~~' ~~o>,o~Lo ~~ww.~ U ~~ aw.~U ~ °>~aw d ~O ~N k~°a~
U , , .
O
> M ,~ '° a) ~ ° c~O
C ~ > M ~ ~ ~.
't," O ' ~ P~ O. ~ ~ . ~ tC N ~
y ~ cLC ø
...-.y N 4-r ~ .p M ~ . ~ ty.~U., N L ~ Q .~ ~ ~ ~ b > H ~1' ~ ~
.'~'.' 4~ ~c~' ~ tip . '1~~' ~ U ~ cd . '~~' U N ~ vi .~: '~ U ~ ~ O ~~
M ~ +..O O M ~ U7 ~ U ~O ~ 'G cd w 1.~., ~ U M S1. q .~ ~ ~'.L7.:~ . ~ t~ p ,.fl .~ V ' af~~~ ~NU"~"
p ~ ~ ~ ~ ~ U° ~ N 'L'3 0~.
?' d ~ + .~ C ~ ,.~ °~,' U Q., c~c ~ v M ~ M
I~ 00 M O~O
p O ~D
. ~
o .
d a~ o N o c , E-~ n~ ~ . r~ :~, ~-; a~
~qxMQ-o W ~~ ~v;. .c~ o 'oo'~, >,C7 ~'~ .
U U ~ q .~ ~ d ~> U ' .~ ~ d ~ ~ '~3 g ~ Gq ~ ~e ocr. ,~~,o~,~"~-G~, O 'L', >o'+' ~ f-'. x' cn M ~ i '~'~,1 :.O U .~, M T ~ b '~" E'~ 00 .f".
M d 'b '~"' ° ~ O ~ . M U of ~, 'G ,~, .:. ~" M 'C 0, O M O
o d o ~-~ ~ ~, on o ~ ~.. .a o U o ~ ~ U
~s o q ~ o ~ .o ~ ~ o a~ O .o ~ . .o a ~ o ~ v °~ od od ~ ~'~d ~U GUS ~ ~ U ~
~ -~~.n~ ~ ~o, o G ~a cd ~a o 0 0 ~ M ~ ~ ~ '~ ~ ~ .9 ~ ~ .U ~ ~ OU p v~ ~ .
'd a MEd., 0 0 0 0 ;~M ~ ~; o o. Via, c ~~ a o°; ~oo o.°oUn' °NW ~ o c ~ ,~~ ~ ~U G~~~~~ ° ~
o . o-o a.p M .
U d ~Ga ~U p..~.~ U~dC7 °~ non~Od~ ~ vU~~~ ~UQ ~d ~ U ~ ~ . ~
T

~~ .~ ~ O ~.. -p ~
~ N N W.~. _~ ~ O
~ ~ U _ O
O c~ U U
~ c ~ ~ ~
~
O

~' ,~
O O U .
U ~ " ~ U
U x U

'~ ~ i.'~. '~''' i.. ~
~ s.~. 0 C ~, ci-' O O ~ N 7.~-W"
~ O

._ ~ O O
cC ._ ~ O
~., '" ' y 'J~"~'' U ..C
~ ~ ~
.
~ ~
.
~
~ ~ ~

" p~ ~ ~
i U
, ' ~
~1 ~
~
Q
~ O ~'Q ~ ~
c. ~
'~" O ~' U

~ .C ~ . ~" O >> QS ..~"r ' N Q N N .
.
' .'3 ~ J ~ ~
..-~ N ..r N N N >> ~~' N
~

aaa a~a.ddl~>Lar~;DCc~,~ ~~x,~U>x~xxr~
r~z b r. i ~:. ~ 4\ U ~ ~ S7 o~ ~ ~p .. , ~ o ww ~

cC x ,-~ ~ ~ ~ c~
~ Er C M
~

~ ~" ..a a.~ ~O p ~ ' O~

x O O

,o ~t > ~ o -~ ~ ~ 3 ~ ~
.

x Ci7 ~
~

U ~ ~
~n.E ~ o.~
o o i ~ o L' o s~ > 'n o o +.~ .~
a~ ~ ~ ~ c~ W n U

,.C ,-7 ~ ~ , o ~ ~ X ~ U O ~ P

~ :~ E-~ >-~o~ ~'~ ~ ~
~ a~ ~ .

c~~x:nc~~f~i>~ >
vU~
O
.

d ~U c ~. ~ p.
~ -~7 U U ~ : o ~o ;..a ~l ~

U O

~ ~ ~~ ~ ...1 Q

U .n ~
x o -~.

~ ~ca ~~x ~o.~.~
~~~b~

+~
w Ys~ d ~ ~U
~' Q
~
~- o E~~o~; cn~.o , .
~-~ ~
~>~.~o~~s ~ Qo.+

U. o oc a y ~ rr o ~ ~ ~.~ oU
~

~_ >,.
, .~
o ~ .n a~ ~q ~ ~s a c ~ ' -~ ~
~ ~ . o~.
:Q c . _ . ~~
o o ~
o ' ~ o~
~
~~
n3 ~ bAc~
U

O~O

O.'~~ ~ .
1.3,~,~QU fn~
G''s.~
F''p N U ~~ ~bD~~ ~ ~~4> Uv ~~+'~-~
~ fl.."y~.b 00,-.,OC~'~vf. y~N~. U+-'N
~ V U
N ,_; O ~
O
U

ct ' T O. ~ L," _w ccf ~ 'y N O ' U O r , :Q ~ ~ ~
n >
U
~ ~p ~O~
' U

.f.' ~
~,Q~~,~:J, p CJ O ~~
U w f~ N rr bD~
"~
W : i U
s .' U c~
U
W I~ 'v~ ~
~

. ., ...
,.
.., .

l~ ~n N

.-. v0 ~ pip ~ ~ .~ .
O
N

,~ N ,n v , '~ ~

W

t~ ~O ,~ v~ ~t l~
O
~

O c' N
~O N ~ '7 - O a1 O
OO m O O v_1 N O O O .-. p 0 ~ pp N ' 0 o ~ '~
~

o I I l 1 l~~v -~~O

~x ~~ zz-xx~~~~

a~ ~ ~ ~O ~ a\ U ~
~ 'O
N

N ~ U a W
~ ~ ~
O N +.~ ~' N .
~ ~
r 0 .E"' ~. .~0.
_ ~
.

~ ~.~e~M01 ~
O ~'L1T3 >, A U s~. M .o Ga o U ~--. 0 0 .D . s~. Q ~ .o U~l.'~

o ~d~ ~'~~'~
~s ~

'~ ~1 ~ . ~ o d ~s ~ .~ Ct" ~ a: ~

.~ 'O M ~ CO C,' '' TJ ~ ~ .r., eC, ~ U ' L
~ ~ o U kf~ ~.b-o ' Q,~
~ ~U ~
U

-yr~ p DU 'n o oE-0:~~j .
a ~ H

~r o o . ~ ;~ 'r; _ ~ ~
~ ~ W .+~ '+~
x ' ~Q~~a ~a~QQy ~a ~
~ ~a U ~, c~ ~ '. ' ~ ,O
y +.. ',r .O ~ ° U ~ ~~ ~
,~ U .~ 'N O
U y i~-. U ty ~ a.. a ~ ~ O .,.U~., N ~ U ~ O y'~., ~n ~ cct ,-~ ~ >>
b ~' ~ y F.~,7.~,b~ U o.
p U , C1. ~ ~ -p ~~: ..C
N ~' O ~ ..-Uu, ~N ~,' U N , ~" ..x ~'c~G~~'' s~...~ V~~aS ~~O ~
~x~U~~~c~ Qa~H ~aa ' cn 4.
a' G ~ . ~ ~ o rn ,o , ~ ~~, d~ .,-, U
o ~ E~ .o o E'' .'~ '~ ~ ~ ;o .u'..' o TAW O V ~C7Q O _~_O ~ ~ ~'~Oa'M ~v' N~>~'~~o Q ~.' O U N ~ U ~ ~ a ~ ~ U V ~ ~, 01 ~ <' U _. cct rn ~-' N c~ ~ V7 "3 . O N >, L U ø, i-U. ~ ..... ' C. d- ~ rUn y .-r V ~ t. ~.. ~ ~i ~ ~~., ~ ~ ,o~p ~ E-~ ~ , ' can ~ ~ c~ U .~ ~...j ~ ~ c~c ~ ,.~ a~ Z ~ d' .
tap ,,°'., =~~ co ~ ''' ~'' Z d d v~ c~ ~ ~ ~ s°'.. ~-°''. ~ ,y--~ ~ ~ ~ ~~' ~ 'W -U-, ~t _ c~
bUQ~--' O p cOG .l~ ~~ ~ ~ C ~~ N O U U ~U ~ U 4i ° ~ ~ ~O ~a . , O ~U U O ~ t- ~ p '~ +~ b~0 ~4 (~ ~ O ~ U 'N~ ~ ~~
cn'+.~ E"" ° ~ .c ~ ~ ,-. ~ ,c~ '.~ o Lp ~ ',~ M t~»' on ~ G~1 ov o ~ ~,.:, ~ ° 4., cG F-~ > ~ ~ ... ~, a~ ~ ,a? o, a~ ~C .~ ~s ~, ~ o. o o 'a .o . ~ o ~ 'o ~ ~ ~ 'o ~ ~ o ~ a" .° ° o 2S d' r~ C H ~ ~U ~ O N '> ~ N ~~r" 'C1 O~ U b ~ ''. '.~, U CCS ~~ ~ U
O(~ ~ CC~~~c~ >'OC,.pNY.~I.cC~ >~...U., ~UtriH~.a~oN~~~ ~-~d ~~~~ ~ ~~ ~ ~ ~Q~
o .~ ~ ~ .
a ~ a ~ ~ :~
=° ~' °' . U ° o ~
o ° ø. ~-d.ov~ o ~ ~ c~i ~~
°: due- ~ . _°= > ~ o ~~ :.° o ° °+~' ~ ~' ~, ~ ~n ~ ~ ~° ~ o ~
U~.O C°'+~UO cc~ '-._C~.~~~'UUL~~
N ~ ~ U O U ' ~ , O 4~ '~' U t>-0 C~,7 P~ <J ~ ~fl "O
> Q:. ~ .- o ~ ~e ~ on -~ . bn ~' ° >
o L.'' Q .~ c~ ~ i " v ~ ~ ~ .~C ~ , ~ s°~. -Un. a> .~ ~
~'s~' ~ ~ cue, N o ~ ~ a7 ~ ~ ~ .~ Ga tn +-. ~.' ~O ~ ~ O ~" ,~ '~" O p ~ 'cue U ~ ,~ '.u > ~ p."tf .t-. at U 'O
M ~ CO
O N M
N .~ ono ., 3 ~
00 "t7 ~. '.~ N b J~ ~ d' ,-~ ,-; M N ;.O M
',~.3. a ~ U ~ O oo .d ,-y N '~ y0 .
.fl ~' oo ~
err, ode ~~,~ ;nUGa~,a.b ~ act >, b ~Us~ °aa:~ ~p:9 ~.flUq~:9 ~ ~~~
o ~ ~'' *' U o ~ ~, ;, o~ E~~,.~" ~ o c ~ o~-o.~ ~ ~ o ~d ~
yt~r~ c~ pQ'~ vi~n~Q c~ ~ boo ." cC
~r ~r ~ ~ ~ o o a~ d~ °' ~; o o :~ tr cn ~
° L p ~,' ~ ~ ,-. ~ .~ ° L~ aS ..p U U O Ia ~~UU3 000 0~ ~ ~~U~~ o 0 oc~Ua ~ .
G-l~ ~ ~'.~ o~~ o~ . ~ ~~~M ~ o o qQ ~ ~ o U~adC'7 ~U~ ~~..., ' UQQ,~ ~ ~~ U~dx 2'7 c ~ E

p ~ N
'O O

C ' C
.

_ +~ ~
~
NO
U
Nb O . .
O
ON

.
c G
p .D .

p ad ~ o a~ ~U~~ ~' , _i ~ '...'c~ ~c~n W~j~
'o~~'~' o , ;~-1~'~,~

._ ~s~oY ~ ~
~~o.c ~. ~ o .o ~ .>
~, +~

~
o ~z a. ~, ~ ~ ~ ' ~s ' ' p +:. O c i ,' ~
C40~~fl ~M"'"'~U
~

y U U ~' ~
~>o O C,' N ,-~. ~ ,.t," X
.~-n 0 U po a-. O z t-~
O ~A . b ~
o ,", ~, E-' ~ ~ d .~ o > o ' G O C
~
~ ~
p G COm _c a , '_~ j, p ~ , p ~ ~ co ~
~

O Y
~ 00 ~ ~ ~ ~ ~ C~1 ~

_ U ~' ~,'OU1~'~0.~.~C7 ~~~
H ~

V ~ ~ ~, ~ V'1 N

rn N
.~ b y ~ S.U.n '~ G7 o ~1 N ~ U ~ ~ ~
~
,' ~
,"
.
.
~ O y .~ Cct Y. .N V~ ~ y O U ~
~ .-. ,1-~~ ' O ccT (~ ,~ ~ .~ f~ '' ~ > aS N
O ~ U X ~"

,~c~~~~U~as~~-o.~,~y>,s.U'a~
A.. '~"..~ p ~
~
~

vi ,~
t..
O y ~ ~ .
~ -p ~ a... ~ ~ ~ O ~ ~
~ >, O .fl ~ *" 4-I GG

O
'n~NQN~~~-'~'OU'~~,'~~N>O~A..
~

o ' '' ~ f~ ~ ~ 0 0 0 o U ,,.x.., .~ ~
"

' ~

s3,U o ' ~
~.~ ~ ~ ~ '~ ~~, ~ a.c.~.~ ~' C
~

O O c ~
Y p p ti .C ,.~ ,~ ,0 O ~' p bU O ,~ ~' ~ ~ a,5 O vi . y U .O x ~.OU~~~pOUf~U~U~C
.O'CNN.''~"..'~c~~N

.
U N i, ~ ~ U L. T~ U ~ O O ~ ~ U
Ti QJ b F"'., ~ "b ~~~~~~0 f~~~p '~~Y
~
~
~
"S
O

., ~
, Ni n r F, U O O ~ v7 ~. of ~ U 1~..~. of O.
v1 ~. C~ O 'C3 ~ U c~ cY b M

O

O d' , O ~ . , ~

cn x3 b Q

bw~

, U ~

O
. ~ c~ .

.

.~ O
p o r~

;

0oa _ oa s~Ua .

~n +:. .
nQ ~~~

U~dd U b0 .~
_N v~

~' U
f1.
O
.~

O ~
~
X
N ~
~
W

~L i ~ ~
~

"pac~~vi~j~~COv7~
L
.
.

~ p"
O .
~' N
'O

f3.
cC
p u. O
b0 O.
O

-O N
N J_'..~
~ N
U r ~ U
O ~
i.~.
dUax~-asav~xa~H

~.,'' ~

,~~ ~
~, ~,~

~, ~ N
~
~ w ~ . as . ~ Q, a ~

cy,~ O c .-U. 7 4: .~ O _p .f"., _N '-.
m ~ -a ~ U a~ _ ' U
O O ~
' ~
~

~
"
~
~ f3.
' O
"C~
N F.' Lr 3~

;~

a. c ~
N
x ~
. ~
_ U
C ~
.
~.
O C
O ~
~
~ ~

x N
,~ ~
'~
'~
U ~
W
x ~
,-..;
~ N
O ~~
~O
a ~.~-U
d "~~
~ ~
~x ~~
U
-a ~,.~
,~
~
o ~
~..
U
' ~~~aSoS~
~~~

,Q
~ .
U
~N
L~ O
'~' p p ~
O

O .~

~ O
c c C
o_0 W 'O
.
,, _, G p ..O

~

7U_~-~TJ.G~_ ~.'0.~
QJ
~
~
~

Cl~vu~
TJ OC, ~ N
l"..
U'OzULLiN

O
O , . ~
7, cC
.
c~

_ ~
' ~
V ~
U p ~

C.' v +, 'b N F
.
~
~
-~ ~
~n~3 '~~U
~~
~

..
~, ~
:D

' 4. a.
,~~
0 ~
U~
~~
~c~,v o N ~
~ O
'O
O .~
O ay p ~n G~
+.C.
.fn ai 7N~~~~O,~c~i..'c~U4.~

~ O
p ' ~''=' ~
~ 7 ~' 0 ~
~ 4~
~

~ ..
~
,~
"
,~.
~
~
x O
~~
~
~

~~p N~
..O~W
b~D~
~~~
~
~ ~
' _.
~ ~
4.
~ O
. '~"~--~' '' O c~
"O p ~ W
'~ ~

n en i .-.
m O
rn O ~"G..
~ '+.
c6 >~
~ O
~
o ~
= 3~
~

.~, ~
~
~
w~ ~
~ o.fl 3 ~
Ca C
~ O
M
'C3 ~~

~
~
~

O
.~ R.
.
.
N N
s., +
~
G
~ ~
v ~
~
7 ~
~ ~c ~
~w ~
U ~

.
c .
U F
,.

C
s .
a, I

a.
z ~w a U
.

o ~
.

~ N
U

o~ni N :
U

' .n U
~' 4~ ,-. a o ~~U o ~> a; as ~ ~xW ~ ° ~ ~.~p~ > ~ ~ ~.?x~
~~ ~a rr.~ r~.~ ~' 0 3 ~ o °Q o,~_~ ~'>
d c ,~ =v o c"°a ov ~ ~ ~ ~ y ~ ~ ~+.. o ~-''' ~ vi z ~ n.
'~ ~i ~ ~ ~ o ~ ~ ~ cn ~ ~. ~ vi .~ ~ .~ x ~ ~~ ~
,..~'7~ ~ ~Q~ ~ en~ c°yli1cy ~~.° ~ ~ ~ ~.C7,~:fl ~ Uw ~ =
:.d "~ '-. !~' ° ~ ~ ~'t C ~ O 1.~. ~ ~O ~ ~ fir ~ ~ '~ ~ i,~~.. -f. ~
C1. U U U
U . o, ,~ _..., of t~ > V, . ~ a~ ~ ~.-, a~ o > a.
+~ O ° tap J, o~ 'x ~ N v1 ~-' ~ .~,.., W > ~ ~ U ' op .~ 'O .,.., U
o ~ ~ ~ --' ccs U~lC7a~ ay ~ ~ c°arn ~~
°°' ~ o o > '" ~ °C o ~ ° aW' o ~..x~''' e;v'~~.
~:~' ~ 3 0,~
Q..N~yi~~'.~HOU~~y~U.~ ~Ny' 'O~17~0~U Q.' Uw o°°.~~xcL,...m-~ ~~°.~v~ ~x p..~ ~
~xxUv~'r.,'H~~~ ~ ~~
° v ~ °. :x C. ~!1 ~.' '~~' 4- U U O , cC ~ O ~ 'O '~ ° ~ ~ R ~ '~ ' , y Q i1 ~ ~ cV~ N O >, ° ~
U c~ c~
~. L ~ o o .d .c s~-~ ~
y Y ~ ~r U ~ ° '~ ~U ~ cct vyn ~ ~..'~ .
by _°? ~ v-, O
v~ ~ ~ ~ ~ >
~s . ~ ~
'O OU N U
.,~., ~ O U U ~ M ~ ~ ~ N ~ .U ~ , ~UQ ~ 0.°.0 ~'.'~UUUwUW p.,°
o o _ M
O
C''t N
tap ~' U
N ~ O
U
U
w ~. ~
>~ .fl U o ,.

WO 01/79442 ~ PCT/USO1/11850 Y
G
~ f3.
x .
s., ~, ~~,x sue. ~ ~ G G
J a' ? °' o 0 'O ~ ~p .U .U
~G L~ ..a ~ ~O "p O Cr ~O CO O ~ ~ 00 d ~ ~ N ~.U.1 ,5-n ~ ""' .~.. O ~ x" ~ , c0 ~ 'i, .c o ~o ~ x ' ., a a~ .~ G ~~ >' °' o i ~ ~'~ '~ ~ ~7 M o ~ ° N
~_ ~ ~ o o'~o~~ '~' ~U °~'' v oa ~ ~ ~ o ~,c~;o°' ~ ~ a~ ~f cn~;

c x~ oY~ ~ ov °~;o ° ~x N4.-. >a Q.,.~ Ha, O b a~ ~ .-1 ~: . ~ ~ E" d 'o ~ G '-"' ~ ~' ~ ,-. 'O d' O c~ 3 aw0 ~ a~
4r .-. G'. W clY ~ Cn C~.' U ~ O O, 0. ~n ~ Wn (~ G ~ ~ "" ~ y.
(-i +.. ~ ~ ~ Y~ O ~ 0 '~' ~O ~ ~ CG O~ U a3 N ,~ ~ .-~ N bA O ~O -fl w U
°
.o c ~ ~ ~ a. ~ on o '"ooa~~~°~o~''~'~'~~ ~ ~°M~~~d~~.°aZi 'G ~ o~ocn~a' ° ~ o EU °cQ ~''~,~~ a. v °~1 ~~
~°°~° a~ ~'ox ~ °'~i ~,~
'"' o w G ~ c ~' ~ .~-.~ ~~ o ' w, oU ao ~rn~ o ~ o , ,> U
o.~ o o..~U o'~_'x'U~~ ~~'~ Gds i G o ~ *' °~' G a ~ ~ ø.~ o~
OGCGOp~G~i'Ot.. V'1ø.G U~ Gc0 aS~nC70.~~'~~~O~G
',.:. ~. o "o Yo .~ d ~ , o (j ~ .0 3 a~ G G G
:~ p-. F ~ ~ ° ° .o a~ ~ M .+, ~ ~ ~ ~ ~ ~ G a: ~ ° a~ a > s1 ø' °Y' '° ~
~yovo G O.~ ~ ~ o U ~~ °~ ~~ ~ ~~~U~.~ ~ ~ ~ i ~~U~n GW o ~ °o, v~(.a ~ >,o,~ o ~ o G ~ ~~ G o~ °~r~~, ~~~~ °c.0'P~~~1 ~
°~.'C~E~r°y °a~~,°c~oo dUU-v and pa.-.fl~,..aUM ~eUOC7 o ~ ~ ~ ~ ~ ~ a - o °' .~ Y o M .~ G U ~ U .~ :° ~' '° .~ R..
~ '~ o O ~' ~° ~' ,.'~c ~ ~ c~ t~. o ~ ~ .c '" ~ m t2. O O U V U 0.
N 4r 0 0 ~ o o ~.~n~ 0 3 ~ o y~ ~ ~ G
G ~ ~ G '~ o ~ :~ ~ ~ '; oo ~ ~ at G Y ~ ~ .
ob o U~ ~ i:,o-I-Ga ~ ~ ~ ~n i a ~ °~~ ~.~ coU
~, uU ~ .h ~ ~ o ~ s~
~'i~...~~.~OpYO.~UG~ ~rUnNOOOH~~~US'r' UU
~"bn~~','~UO~U~U~v7p 0,~'~,.,~.~~,.'ptcSU~OT7~ N Vof O
O ~ N ~ . aS ~ :.d YO. v Q Q. v~ ;.~ k v~ y'~., ~ ~ " ~ 4, ~
q~-o~'~U~~~OU.~E.~~Uo E~-~.~~°.~°.~~OU°°~.~~.°°ov°
M
l~ N ~D

N
O O ~

U
I
~a . ~
G
dw '~G ~ ~ °~ M .Y o0 U ~ ' ° o"-'NdO
V ~ N
~D N . ~ ~ ~, -~~ 00 ,.OT
0o cct a ~ DC o Ca. ~ .~, U ~ ° ~ ~ ~
o ~ d ~ -o ~
' ~ O (V ~ .~ ccf ~~j O ~ ~
-fl U o C ~ o ~Ud~Ud~~~ ~U ' ~
"' ~
~

O
VI
QY
..C
Y

>1 y w O
N
~
p ,."
~
N
~?~
oa.~~., ~.~.~-O

_ ~ ~~n~
x~
~~x ~~

o .
' O
~
Q.
N
L~
U
a.
~
.~
-p ~
~

~~
~
~~~b 0~

an c a ~~i~~H~~
~HOd~a r U N
a ~.1 w 'O
.-1 O
~' , , ~3z ~
Q

~;o~x~ ~,,'~or., ~~
~~
.
~
~~

w ~ ~ ~o -o ~
~;-.o~~,:.~
>,o ~.
~
x ~~
.

~
~
~~a ~
~
~

zs O ~ . a.~-oE'4-~..~
+' ' i ~W~
~W
..
.' d N
U
~
f3.
T
N

~ U
.y, p +~ C/~
U ~ c S
p., a '~
--~
U
_'~s o ~
..~
c\i C ~ ' ~' U U
fl GL
C V
~ ~L
~ O 27 ' . ~r~~n, . ~ Qf~~t~E-.~dv~
c m C
~O~~cv'as..

vi oo ~ fn d" y-.-.
.~, p w0 C W ~
~. W o . ~ i r~ ~ . ~
~ ~ d ~, Q bn , ~~~~~o~
~> . a ~U , c ~~ .

~ .
, ,~
~ o~ ~~
;~ -o ~ ~
E-.
~
a~
~
R"
o ~
-~ , on~:x ~ ~ ~
~~ o U~..~d ~,~~:.~.
U
~
o, 70U~~Nm .
N ~Urnp~N~~'c~'L."C~bO~
v1 ~
, O
l , ~
c0 i. ,~ ,_ ~
.7 Ga ~ ~ , ~ d at ,b ~
~
C.
~p ~
.U
'b N
O
E
b U
.+~
V

~
O
.
p ~~~~~~
~
~

w .
a Oza ~ 4. c00~
>
V
~d ~
~
V
N

dppp Q C y O .~ ~ [~-m~C
~ ~
~
.~
by ~

~
~~'' '.~~ 't o o R y ~ ~ ~
O c~Cn-a N
.~
.-.~
~".r Q
U
~' U
~
~
O
U
fn f c ~. pp O b O +, .
~DG~ .
"y i y~ G
' ue ~O~N..-..
~~.0~-' ~, ~' ~

._ ~ ~
. , pp~ ~
V V
q n.v'_"
~N
~~t ~
~
U

U N O G4 ~ ..flp -r". .d 'O .~',~
~
U
' y ~
~' vW"~
.C1 ~
~
't ~
o w,~o-o ~

-~

O ~ ~ ~ c~i''U
J O ~ V 4r ' ~
- bUA~
U
~
ai H
~
E-' ~
~
pp~
~' ~
~
U
O

~.~ o ~ ~ ~, .
~,~.~ ~ .~
~ ~, ~
~
~
~~~e~
~
~,o a.~w ~~
U

d N~UO~+~..N U= d'NO_~.fl~~~"OyC~,~~.~[a-r~i~.,'f-.'~'~..'d.~..pOb.

~ ~ ~
~ >

. ...+.
O ~.. ~ .~ N
Ga ~ ~
~n c~~-r~ Q.
." vy ~
N ~
U
'n ~
~
r+~n a'' ~
ca o +..
oo H

~
(~
U
C
>
' "

~ N +. v1 ~, 00 O , U
" ~ t-. >
V ~ N ~ O ".,7 ,~ U
sp. ~ p '~ c , ..
N ~ ~
'O ~ ~ ~
N O U
E~' ,.~
~ti V
~ ,~ ~
~ ~ ~
G ~~ a O
"p E-~ ~ .x ~ ~ U ~
U ra. U ~ E-~c E-~ .~
C7 ca U .~
U c~ ~
U o. >
U r~.
~ ~ ~
~ d 3 c~

N

Q ~ N
cn , O

~~

a, Lp O

w a7 E'-~
a ~-~' a~

O C

.
n a~
, ~
U

R o O
~

,-O U ~
~

N d' p p O

00 by ~. b rr ~' ~,' Wit' O O S~
C~
a a ~

y .

'o ' x an an ~.
.:
.c ~i r o ~
o~
.

~N

w ~
N

.~ ' ~, cC

:.d k ~

p T

~

V
'L~
..*'.r y O

~

O
U

'C

O
U
~
U

U
~
~d ,:~
O

n n ~ ~ b ,bA
T 'L3 .o~ ~~. ~ 3 .a~. x.
x 0 ~

a~q ~:~ ~"
~
. ?_~
p ~~

' 7, N
-~', ~ ~
i ..0~~ .~0 'O U
~
'~~' CV.D
~
' ~ 0 .fl 1 ~G
_a~

. p~~ ~r~.
0'V ~ o~o ~N
~
pO

p~~

-a x -o x .
;-x G~
~
~
'~n ~o .~
:
'a .c :~
~1 .c ~~
o ' "
w U

~' b ~
.
U
~
O

'k~

~
~
~'.~
o ~.
o 'b ~o ~

~
.
J
E

o ~
as ~
.~n , i ~~
o~

:b .
a,.~

x_ x x ~

;
n~~
~n ~
a u ~
o n.
~d~
~b~

U
C
U
o ~ ~ ~
o v, eo0 0 ~~,'~ ~ x o~.
''=' ~-~ E-a C~ '' x ,--.
o ~'~ ~~ ~ as'".~ . Uw y+ ~,~U o .b ° o ~° ~ ~ ~ x .-, W o ~ ~ ~ ~ = ° ~s N 'b ~ ,~ ~ p~ '~
~~j ~ s~,NC~tar, ° r ~,~+- ~
+j G0 ~ ~ ~ ~,j ~cr c ;D ~ -° ~ ~ .N
. . W v7 ~~~.°~ H-n~7H
.~-°~~ ~ ~.~~j v o~' o~n~ ~~ ow ~ ~
° ~-o.,';~ ca U ~, c~ ~
~~ ~Q°~ua~-u ° .r' .o~ '~ °' .
o ~c ~. ~ ~, :~ y ~ ~ ~ ~a, C4 ,~ ,c~~a O ~ ~ ~ 'N ~ .~'n. ~ ~ ~ N
U pp ~ U
~ U cG ~
w.~ o.~~FGr'~U'c~~'t~ ~;~.~ 3 ~~~P.UUvo c a ~, .c ~ ~s 'n ~ ' i ° ~ o .C ...gin. L: N -~ 4"' ~ cC O y N N v~
_~~ ~ ~._pU_~ , ~~ N ~ ~O N.Wn~ø'Cd/70 UUaITO~~.~cCSS'"~" ~'~'' ~~Tj t~ NN~~,r~U .G U
.~f1"U~N~~ c~C ~30W O ' O O~.D... ~C ."f'~'x~ U
J, by ~n ~ ~ 4 ~ ..O ~ n ~ ~n Q ~ at ~ ~ j N ~ .O
c~~~ ~~~+; '~ a. s'~ o ~~'~, > a o o ~ ~
V ~ ~ ~ ~ N ~ ~ O O. ~.'~ Q ' ~ %~ N sØ ~ ~ ~ cG O ~ ,y~-O. O ~ ~ U N .C.'' ~.~ ~ o ~.~v'o~ ~ ~'~ ~C7 c 3 ~ ~ ~ o ~ °b ~,n cCt~.,.U,i..,N~p~ '~r"i~~U~ ~~~r.;~Q,'OU4~ ,~~~NtnN Cw'cUCy., O +. ~ ~ ~ i.. ~ ~O S~. ~ ~.-. ~' O U y') O .C ~ O .O O ~ ~ t» O .O~ U N
o v H w. ~a a~
~°'~"~°~~ao,bn~va.a.~~+ °o.~~~~~o i3~.o~~~w''~.n°

~r ~, vo v~
N O
O O
O\ O
O~ N

y .
a, N
O, .
~ O
'~ ~
U
U
O l~ ai O
..o W 'v 4:. ° ~ p , tf., .C U ~,.
O ''' ~ O ~
t"..,." O . ~
'~"' N . N ~ ~ , .d ~ O O
W ~ O Q ~

°
U
N N N cOG N
U i.: U
U ~ ~ a U U

v i ~
~ ~ a. ~ . ~., o ~:° ~ o ~ ~ ~ U
o c~wv°~ a.UaUOw 0 0 o~

° °" . ~..: oN ~ o ° c+.. . ~~; ~r ~ o ..dc.,O~ p~rs.. G r aS ~ ° b 'W ..,Op text.., ~N~ ~ O
.~Ni='O~s,~.~ ~~~~' O ~W ~Oa;~~.0 ~~x-~ G
:bo~y°'~~?pHEn~~~ ~ ~'~~~.~w~~E-.~~n~o~ a~
~ b~n'~'Y~'~~ ~ ~ o~
a~ ~ d -d >. o ~ °? '° ~ ~ ~ b° o ~ ° a~ ~ ~ .~ >;
o ~ ~ -° ~ 'r ~r by o ~ o ~mo~~ o ~ °~°a ~ a~ a~~~ ° '~ o ~~~ °.n ~ ~a b°a ~ ~ ~v o °° ~ .'-' ~ ~ cn C7 ~ ~' ~ ~ .° c~ r~ C~ °' ~o~° ~~'_~~~°'~U~o~o' ~~-ono .Q x x C~ '...' o~ ~ ~ .°' '..~ p~ ~ v' d: k ~ x ~, '.'" ~ C ~ ~ 0.i N 'r ~ O ~ L T7 s. ~ .~'...J J, ~ ~ V ,,.,,0 O~ s~Ø. ,Zf O ~ ~ .~ ~, s.. ~ a~ ~ "d L~' m ~ ~ ~ ~ .~ ° z G
~O ~' m -o >, v _a~ O ~ ,~ O ._~ ~ .~ N T7 >, U ~ ~
.d . ~~~ ~ O ~ ,NA~C. +' ° ~ N N ~ -~ v ~.~ U N ~ ~ ~' ~O N ~
O~~a'~.~~~~~.'~~°U'C. °7C~ ~ ~~'. ._~~C~°UT3 Q U ~ U U c~ r~.~ V7 ~ L~ '-G 'O U Q d ~ ~' ~ ~ N ~ N ~-, U7 ~ E.. .y~ "Cy U Q
U ~ ~ U U c~
° i.., ~ cC r.
N ~ o E..., ~, -t- ~ C ~, . .t".. -j- U ~
~ U by 7, ~ ~ .'.~.' ~ 'O p ~~ N ~C~ .
~° .f,~.'~Ov7U~OUU ~UO~~~UU~"C7~O
o ~~,o~~ o o ~ ~ ~,~ ~3 ~,~a c~ ~,:~ ~~o 0 c~ ~ ,.~ ~ w~ _... ,.~ ~ o c~ ~ o a~ ~ ;r ° o U ,., ,~ ~ i ~ o a~
O O ~ O ,.C N f3. O ~ ~ p ~N ~ y~ ~ ~ ~ a3 ,~ U N ~O ~ ~ Wn a U o ~ ~ W o ~ ~ ~ ~, ~ a U t~: o Wbp~vanW~.~~WC~va~, W~ i~~~.~~WGav cn, N
~ ~O ~
~m N G1 I~
cn ~ M
W W
DD ~
V7 Ch l° Cn -tn-n O
i °
~z ~o W cp"'.7 ,~° m~ ° .o .~W ~oUX W
b°noo_o.:...~ o~.o ~~.~pd'_, QUO ' ° Q
b U
0 0 ~° ~ ~ ° a. ...
~. ~ W ° ~ ~ ~ ..n W ;'r~
w~awa'~ w~QQ

_. ° O :r as ~ i.; ~ .
a~
~; coca c c ~ ~ ' O C~ N ~ cc3 cc1 U ' w.
U U Z o U U ~ ~
~ ~s'°z ~.°UO - ~ ~°.
~"' O ~ .~ - - ~T ca vl i.. .a U ~ s" +' U U ' ~ ~ U
~ cC q .-' C O ~ O ~ U ~
c. O a) as N
f~UxUU0.~p;UU ' ~~U
x ~ a~ v~, ~, ~ ~ ~
~ ~ ~ :fl ~ w ;~ ~ E y >
0 0 ~ ;=', ' ;
U~ ~' ' ~ ~, ~°'H~x cU
°o ~~'~ a.~~~.~x 3 °.~
~~ H '~-i ~ .~ V CV O. .rf, ~ o '~
p.,~~ p,ON Ny~~ O~~N
oN ~~ o ~ ~~.~,wd ~ o >=1. N ~y, a7 "~
'~~.
c~ N p~ '"Owd~'+~ c~C,W-~» c~C"~~~
O' ~ UC'.. °~N~~~r-.~~~Z,~1 3 ~ _ O CV ' ' 'O J"., TJ ~ ~ >,'t"-. .~~' E~
.'~.."U~~UOU~ObII.Y~
+~
w v can Pa w w° w ~ ~ b x ~ v~ °~
>~ . .~ , a~ a~ ~. . o ..° i w s?' ° C G ~" O
~~_~ ~ ~ ~ ~ 3 b°n~,~;~~
W ~ ° ~ . '~ o s'~. ~ a~
y O .~ V
.~.~ ~ '~ rn O ~
~"~..~"'.n , ~.upp~p~pQ.~~~O
4~
° c~ ~ .o ~ 3 , a~ o ~ a w on ~
v c~ +° +J . ~ ~ ~ cct i w ~ c~ o ~ o~n~ ~ ~ .~ .b E
,., . vy . b~D "~ '+j ~~ c~ . w '.O +.. N N ' ~, U
a~ o.o-- i ° ~~ ~ ~.~:~ o'b a o H 'c? ~ ~ ~ ~ w~~'i ~ c~
00 .-. o - o~0 0 ~t u-, ~
~ ~.
N ~ O~ oo a~ .-. ,--~ c~
~n N r ' O ~ vW t '-. ' .-. ~..
cal O O c~ 00 00 ~ O O O~ - CV ~D
° I I~ , ~a~z~~ ~~ .
M ~ :., ~ M
_a f~ ~ °_c%, o ~ o ~.~,.~>,o.c owl d....d~
Pw tn U ~ 4~ ' J ~''° ~..' U w t~' ° OU ~ ~_ U' '....~ b- Q
N ~, rn ~ O
cd ~' N '~ cC O y ~ C ~ C [.QL] N U7 ~ ~ ~ ~ O ~ V1 ~ ~ ~O 'C cC a3, V O
o U as w ~ oW~d ~ ~ a.~<Co E>C '~.b~,W ~ ~ ~ ° ~N ~~~.b~ ow >,p -c ~ C7 0 .~ ° iii o t'~' (~ o ~ o ~ ~ w o ~ ~ ~ .~ ~ ~x U ~ ~ °
~ o ~ o E
~3wW pd ~c~t o ~.-"~° ~~~dC7~~ ori'E"'~ ~~~''' wd~ o ~~~°~NasU~o..N~oGW~W~~oW~vU~;'a °c~ic~ao~ . ~~°°~
~ o ~ a w Q x ~ ~ "' ict U . ~s ~' U
~.flW o ~~W o o ~~V'a~ni ~ ~ ~aboW U~~ ;o '~ ~ °d o ~d o ~ o>xE-~~.;~° ~ww~"~ww,:fl"' N C *'' o +~ ~,:, ~ w ~ ~ .+. 'D O W +~ *.' ~ c.t ~ O ~
W r~ Q, d ~ ~ U Q ~ ~ ~ W ~ ~ ~ W ~ W W ~ ~ d W ~ U w Q w ~ w d U ~ U cC
.
.

4 bb U .cC
~' .U .U

o ~ ~ Q o a o oa ~v o :o~ ~ ~,~~ U
~~

~ U
O O ,~
~
~
H
~

.dx~ cn x .n ' a , o'o ~ ~ o bn d z>

~ c> ~ vi ~ i on iC7 ~" ~.~ ~ ~'..fl o ~ o~

a "" ~ ~ ._. ~ o a~ ~ ~
~ U ~ n ~ o0 ~"

r~ ;' ~ C'~
~

o ~ ;..7 d w,~ C ~ o U -' an >
~ o c 0 o ~ -b , .
o , fx ~ ~ ~ N
'~

:d w ~ te ~
p ~ ~ w ~ ~ 0 3 ~ ~ ~ ~
' a~
~ o ~r _ a _ _ , ~U
, ..-~ ~
'' ~
O ~
~"UlV '~
~
7~

O ,~ .
~ M . .
d' U U ~ ~ ~~> ~a ~ ~
O
~ ~ ~c4 c ~ ~

O U e.. (~ . .~ ~ 4.a k a~ ~ ~ ~ 'N ~ .,~ +~ O (.~
' a~ ~
~
~
' ~ ~
,1-. U.0 M ~U.~
~ W
cl~ ,~' O'.~ U ,f", n. 7-.
~

~ U N ~ ~' 'O .~
O ~ ~ ~'~'.~ ~ ~ Q
~ ~~pGl ~ N
O

. N U .
. '." ~ ~' U~" ~ O T
_ t ' ~ ..N-mn U V O aj ~ ; (, ~ CU.7 , ~

'Of.' ~~ ~~~ - .~
~ V ~~~~.CUU~ ~~~~V_ NN~
O O >, U ~ .
Q

w ~ ~ O ~ O
~~~ ~C71z7~~ La~'~~ ~ -~ > vU Ud ; ~
~:
~

. a a , c ~
.

-d a~

U ~ x ' o ~ ~

o ~ ... ~ ~ a.
c'~

U

n>

, , 4C7a' '~b ~
"
z~

~
c b~.b ~

a U $
~ W
o c = ~
. o ~ '~ i i o ~ o ~ '~

w G o ~' Ga ~
a o' .o ~ ~' o ~

~ o ~ y ~ . '~..1' ~ u; a~ ~ 'b ~>
r-, ~ b4 ~ U b0 O ~ >>'~ U
~

4a cct .~",.,.. .C ~ b N +~ 4, O O ~, c'~~7 ~ 'C3. ~ .~ ~ L, U ~ i. 'G
c ~ 3 -' ~
~ '~

,~ ~ . a~ o ~ w, o b >, o ~ >, ~. E-' o " a. ~. i~ ~
, ~ 'n ~

~,~.n~w,(7~U', .~
. ~~~.~,~C.o cUG ~Tj.~j N~ ~.~..dbA -~~.cC~'O U N U
~., 7, U O v7 w ~,' ~.-. _ _ _ ,~
~ ~ ~ rn ~ 2s -a ~ ~ O c7 ~. > nn .~ ~ ~ U ~.' s~. w i~ ~G V7 01 m N

N

m ~ O
, a1 01 3 ~

N
C\

N ' c d\ ,-.
n a O GO
d cn tn N O O
~

a ww '~ .~, -Q M

'b >

.
>
N

U
b v7 O

v U U
~
~
x U o. ~ ~ c c ... q ' O

w ~ c7 U ~
bnx ~d o oW ~ o~_~o, Y ~ , 'CS O
O .

N O . p.. X O O
~w~ ~ ~,~ a.E

U ~.~ c'9a Ll ~A >,~ ~~ ~ .

wQ ~~ c~QC7 - c~.~Q bn->

c c ~ ' ~

o ~~~;~ ~ .
~~.~a ~
.
Q

~
o on ~ o c U ,., N
U

.
~N..N.~OWn J ~ 7~, 'C

~
~ N
U
~1 N ~ y ~
~

. _ C
U
.~
O dp 0 '''' '~ ~ '~

C [n v ~~Q~
a ~~~'~aH

c r ~ ~ 3 s~
~ i~ ~ ~
:

~.
, x .~ Q ~, ,"_, a~
~ a ~y ~~~~ '~
c ~~ x c M
~
~ ~~~ ~

~ .-~
~ W ~ ~, ~
. ~ ~
c r ~ .~ ~ N ~, s~.~
;~ U ~ ~ +~
~.
s.

_ U
_ +~.. ~ n N 7, ~ U p O ~ ~ .~ O
~ O O W--' ~ ~
~ > ~., ~
~

d0 M -. ' y. ~
U O ~~y G' ~ ~ O c~
~ pp ,.~ N N ~--W ~
'O ~ 'L.~' O s.

_ ~
o ~ o'~ ~~~U o~ f..1 ~~~~ ~ ~.~ O ~
b C

: ~,,.., .
n U ~ cC
bD N ~ .O , ~ by cF,, ~ U ~' ~ ~., ~ O C/~ O ' ~ O ~ ~ a -, ~ *-' V ' p ~

ai I~ ~ N ~ ~ p ,..
[/j C ~ O G U ~ O ~ .
;~ a.. y=. L~~, CIA a7 ~ O ~O O
~ ~
~ '~'~ ~ O

~ _ ~ U ,r ~ en r _ ~ O
y ~ .~ cn .
e-1 ~
n N ~
~ ~ u. .~ ~
O p due' ~O cC
N

, p ~ ~ ..C N
. s. ~O by ~.~", .,~ O ~"k + 'b ~ ~ cC v0 ..-. N > ~ ~ q -d O.O~~~~y.0U7'.-C zU~'G,,~~UC~%~
~c~~~p~y~~O w U r'~ ~ ' ~
~ ~ ~
o ~, 'r' ~ ~
W ~n ri ~ . ~
~~a~a~ ' ~~
~
~

N Y
4 > ~ ~ pip at y-~..~_~ O O ~ '~
c. t-~ ~. ~ ~ '., c~ ,.~~
~ ~ V CV
~
O
X

~ ~ N U .fl ~ ~" N ,~ ~, :..~'~.
~ O ,~N O ~, ~ , 0 i.
U T
'.,=, J '.i". a) ~ t.~., e0 O, .~

O p ~O 'O Q..-ty _ ~,4?
V p ~ .~ ~ 4~ ~ ~ ~ CO v0 ~ U .C ~ N
~~i C/~ .
'" X
~ ' ~ Q
~ ' , ~ L Y
. + .. ~
.a.~ .! ~ TSriW
- ~ ~
.O a., ~ T~ V U wz ~ ~U~E-~
V r.. ~ ~ > ~
~U ~ 4:~ Q
~v~ ~cnw~~Gq~ ~
~ o ., ~
.

~ n.~ ~b e o n _ ~ ,o 0 ~ a ~ ~ ~ i ~

~. ~.. ~s ~ ~ ~ ~ ~

C ~.~ ~,~o ~ ~

~.. ~ .~ ~ '~~' N ti ~" ~
~
.~

o ~.oa ~.~
~

~ ~ W O ~ ~ ~ U

b~ ~, ~"'~ U
- ~ ~ ~ ~
~

o-- O i, -y , tn O.
1 ~
~ CO

~
.C .,fl C~
~
~
O
~

.. O
~' v- ~ N
c O c , +
C
U

10,0 M
O~

O

~O d' O d' M ~' N

_~ N
. .t". M cYC . N

~ ~
~~. ~

Q, ~ ~D
_c cC .fl cct ' a . ~ ~ .
.k ~ ~
t. (~

dv~w~, ~

N

O ~ ~~ C7 I~ O ~~~r77 ~~

(yes O NM~'1~ O
O E-' ~ U' 'fl "C3 P

~ N ~
O o--~ ; w -n ~~ ~~~~o~z , add WO 01/79442 . PCT/USO1/11850 -o '~ ~' o ~ ~ o' ' bra ~~. O
~v~U

o c ov r~3 ~ > ~
~ ~ >
~ ~N
~'~3 ~ ~
~ o ~, ~~'b:~ ~ o 3 ~ ~
~ ~
.W.;

>r _ ~s ~~.N o~~ ~ ~ oz ~ N

_ : O O 7 ~ U ~
~ ~-' ~ ~ bU
~ p N .
~" :y y ~
U ~

d, . ~ ~
, .. d U _ ~
a. _ . c s ~E-"
a ' 'i " ~ 'o o ~ ~ ~ ~'~U
-cx U' ~ ~ .
~
U

.~ U ~, ?~ ~ S1 U O~ i _U ~ ~, x ' .. "C3 > . U
~ C~ -O d TJ ~ t~
~ 7 ~

~ cti Q , T
O\ U ~ , ~ ov .a G ~ ~n . 3 O ~ r. ~ ~ O~
~ 'an ~ ~ '~
~. -fl U
'r '~
.~
x c c ~.~s o o ~. U ~
"~ o ~ ~ o~l r,., ~ u..-.. ~ ~ a~ "
~ a ~

O ... a~ N O
N ~ Q '~ C ~ ~~ O
N [n ~ :.O O
0 >, v '~" ~
O P..n .-.
~
~--1 U

. .n ~
0 ~~ .
o ~ ~ ~
~ ~ off, ~ U

, y _ . .~ N ,,o, +
. a. '-' ~ o ~; ~ ~ ~.' o H o ~ o ~ ;~ .b a~
.a~' W CG ' o fl _ 'n .' ' ~~~~'~ ~:-~.o ~ba'~'~'~ +. ~r o ~j 0 '"-~ ~ ~' ~ w x C~
~ o ~7 ,..1 o o a~ , ~.-, C f~ ~ S x a~ ~
W U c .~ t~. U ~ ~
~ : W ~ ~ U
c~
bn CJ O ~ .
= ai 'W' ' "p .p ~ s.. b0 U
U
a) bD,7_ ~, >
w N-~
-~
-~

~

~.~
Ox ~~~~xo,~o ~

O fn ~ ~ ~ V
~ O ~ ~ O
~
V
~
O
O
.r.",.' at c Gcd ~ ~ U
~ a 'O
~ ,~'r ~t -~

. .,.
c~ ~
~. -O , 4 ~e .~
.~ +:~
~ ~.,' ~
N
r"
oU
~;
::
~
o o ~ ~ ~ ~
~.~ ~
> ~
w 7 .~, ~
~ .a ~ ~ ry .
C'r..
zr V
' ' .~
'~
Q
~
~
.
O

U ~
~ .p.~ 4 +, ~ .C .~ .
O O . '~
.t'~'.
.'..' ~
bDO
ca ~
O
a~
.
x f_1 . n N , i-' ..O t7 r O O ~
N
O

O . .~. ~ .O
O ~ i x N
A -~ ~ . 'C1 U V +~.' ce'' U 'Cl ~
O ~ ~ 'd ~
~ N ~ O

bp . . , bA _ i ~ ~.,.~( c - O N ..C O' U ~ "..
C -. ,~ C cOe U ~~ C N y ~' ~~E~~-' ~>,d' j ....,.3>~
~
"
~x ~~
o ~
~
~

~~..~~~b~~~.~ x ax~rx~aoH.~~~~,'~~

~~

w 33 ~n N

N O
~

M
~.~

O

O ~ O
,~
~
~
_ vU N
b4dx r-.
N
A"
~

b D
~h wU';'~

~

p., ~
O
.

~ ~n bA
~ ..O . O
~O
~
r, ~
~
~
"~
~

<J' N
U
PH
V'1 .
, U
o.6C7.~

opo o-ooU

c ~ '-' .
of .

a b17 ~ U
~

bD
v1' bA

39, a.

c o >_ x UP~O
~s v~ ~ ~ o ~ o ~ o U O
+r t~. > v~
~'~~~~~y~01 ~.,~ V7 ~. . d ~ w '~ b~ ~O iC U
+-~
O
Nbl1 ~ ~ ~~ . ° ~ H ~ ~ ,~ cN ~~ O
o.a ~dx ~ x °'N °n~'~ a.~...-.
od ~ ~ o~.~ a~~~ o,U o N cC
o ~ v»n ~ " ~ ~ ~ ~ o u. ~
~" ~ F~' .N .b Y X., as ~ o a~ ~ p s: ~ a~
Uf~ G'c~ .:L~O~UO-~~:~~ S.." ~C'.,~ ~~O ~~NNO ~c~
~' ~.fl ~ N ~ +J.~~~L ~~ c~~ V O p,.~ ,-1;.~' ~ r.~.~~~ U ~.C
Oc~'C7~~~',~~,"~~~ .~"~~c~G~~.O'~x~~~~N c~i-,~~~~wUbp,-,'N.., ..-~ ..~ N U O N y" yU" ~ GL ~ ~fl ~ aS '-~' w ~ .N Q.'~,~ O ~ O ~'C1 .~ u~. U
GL . .r'.,;1 .,,~ O N ~ ~ O ~ O N -~' . N t~ U O .(~., U U i.. ~N" s'-' b4~.'O~.'bA~OOU~OO~~N.~.7U'...~"" .yO.,OO O-W>,v~xNOOC,..,.~
_~ ,-. N ~ c~ M ~ >, N ~ ' O o.. N TS ~ t~0 y O
'ir c~,'' .,-,° V ,.~ U ø..fl ...>.~ ~ U .y"', ~ f.~. U 'G O ~ ~, O CG
~,~,, ~ O 7..
O ~ s:' CY ''~ ~ ~ N +J l3. U ~ p N ~ O M ~ L N .fl ~ .0 4: ø i.'' .per ~" N
v~
O .~ ~ ~JF'. "t~ by ~~ ~ .~ cC O .4: ' U f~» ' O t~ ,.~.,, '''' ~ T3 N 'n Y . ~ ~ ,.Ur r~ ~ ':~ ~ ~' '~ ~ O O U ~ O ~-1 N ~, N ...~.. 'O ~ U ~i.~ O O .~
'i.~ ~, ~ i.UJ
M . ~~O~l1-O'~~~MQ..~OpO"CZ,+.C. ~.,U UOc~~N~~~O"S7,U
~~O,.~cxG~~:~~~~O~~NO~NM~~~'~~cn t~.>~~4~-r~~~W
E-~ . c~. 3 ~ ~ 3 ~ ~ ~ ~ ~ ~ ~ .o ~ ~ ~ ~3 ~ ~~ ,.°'. C7 .~ o a.2: o .~ ~ -d -cs m .~
M O~ N
O ~ ~ ~t N. N - O O~
C~l ~f' ' . v0 M ..-. . . c0 V1 N M ' , ~ ~t C~1 ~O c'1 ~ ~
O~ CO t!1 O
_. ' a~

~
~ U
M
~j ~ M o °
by ..o 0 o a~ d . o °
M ~ U N f U OM
O.f.~.'r-' ~ _'Orr O .fl O b O ;fl ,.~-.~ C .~-.~ ~l. . ~ ...'..' , . _ .
O
b~0 a .b~~A O b~A a d E-.n > cn o ~ ~ ~ ~ ~
N ~D U p ~> n ,= ~ v ~ ~ ~ U '-' c~ ,~ a o. vo ~

N
N
~
~
~

N
O p .O C1r c C
N U N O WO x ,~> *' '~:.
ran N
' ~

O .~. V1 V
U ~
~ b L~ ~
a U

~
6'., ~
C, U
N .
U~.'. ~ T ~.~N ~

O.
C ~~~, ~n ~ ~ P., ,~
~ ~' ,= 4-:

> ~O N ~ ~ N ~ V '~ x C
N i. .

CCUO~> p?, Q" ~
cOCCGOc~C'.~

1-~ ~., >
r ~
.~
---a _ x r t-O_ ~
r ~
r ~~
~r~c~~~ ~~ a~
W

a S
~ O
'~N
~

UO

G C
U C
. y N
,xN~.~cOCy~O~Nt~.:,~Ov~7 _ Z cC .~ ~ '~ C ~ U O
O ~~ U T E O

~~~,~ ~~~~xaa~, ~ ~~~~

N ~ U ~ GW ~G a' O ~ ti ' o ~..
N +.. ~
.o ~~ > o , a, ~; ~ o O ~ b 4~"
N
O

~ ~
, .. "~
'.~.b ~ o ~

~. ~a :o .~
c > ~ O a, ~ ~

y a n' o ~ c ~.

b ~ .o ~ c~.
~ C~

a~ o ~ U W

y..i ~" ~1' ~ ~ ' ct F".
'b ~" i.U-~
N U

O "~ x N
~ O O
CJ ~
~ ~

. N
. .

d' N d~w O 'd o ~

a1 cn~~o~o~

O ~

~3 3333 ~ Q\

C~1 N a d xx a o .b a G~. ~-.. . > ~ U
~

N ~.., O
O ~ O ~.

1,. W N
y i1 O

b O N n N <h O ~,.~ .d 'G () -p Cp a~ O ' ~ cV

. U "p ~
x '~'~ x ' Lr +~ W ~i ~i x "a °
_ U
C
c~ c~ i-° i.," ~
U ~ U U U
N , m ~ U of ~ ~ ~
~: i: ~: ~ c ~ a~ ~ 0 0 ° .c .c U' U U a ~ i:. V ~ .' "~' O. fl.
~cpCC~CN7~0'-' ' U U U U O a, a, ci a Nx ~~~° ~
.> ~ m '.°. ~ oo .yn .
~ ~vr LO ~,''~
rp ~ U O ]~v U 13.p ~ N .-'~
U ~ O U' ~ V'1 '~,N
N '-~. C~ ~ U "p,.4~ .r.. v0 U ~ bD O~, vs s.~~~ c~a O ~-~, c~ ~ :O 7 W 4-n ~ ~ ~ O
U U ~ 'C cC Z
'~ o cC by U ~ ~ O
~ ~ ~ U ~ Gt. ~ ~ o o. ~~, . :~ cwt ~N
_ .-' .5 0 ~ ~ o ~o ~ ~ x. ~ ~ ~ ~
~ o y ~ oann ~ c°~t ~
a; c'~c ..o ~ ~ d U ono W H °' ~U Wo U - ~'., ~ w ~ U ~ as O O vi 0~~~ ~ ~a~v'w.'~r' '~NCv~7~U -_ ~~ccS,D~UO~O~U
O W ~ ~~ 'n 'n "p ~ U ~ S7 ~ O ~ c~ xbp p, ~
'b~ ~'0~~~~~~ - ~,~0~,~ T3~nN xø, c~C ~ N b~U N ,~ N ~ ~ .ø, .""'- "U ~ p ~ U x , .~ f.. .~O~, ~M ~ ~, .~ O b0~p ~ +~.,.'f~' ct-~G 2~. ~'~-' ~ O~~'.O~x.~ ~ U ' S3. O f..' 4.-n ø,'y., r".. T U U Cue,' ?a ~ ° ~ N N V
p O ' N p U ~ O O 'y U U ~ O ....
d ate, a,3 3 cH on~.~ O ° ~ a.o ~~°~ ~ ~,° ~ o N t~ o0 ~O V1 D\
00 M l~ ' ~ d- O
N ~ N
~n O N -a1 ~ ~
O O O .

,:-" o .-, > ~ U
o; ~ ~' ' w .b .,~ '~ y b ~ o ~ v ~ ~ ,.., o '° ~t a, c~ ' >, a ' ~ ~ o ø, ~ o ° a, co vo ,~ °° ,-7 ~'~ ~ ~n o ~ ca ~ av on s.. W tz7 .-.
00 0 os cn r-:
o °
i~ ~ +-. ~ ~ a> _. O
O N V ~ °a ~ C N ~ >, ;.~ ".C = ~ , 00 _G.1 ~r oo ~o N M . , rx ~ c o ~ ~ ~ ° ~ o o ~a '~ '~ v, o oy ~j ..c ~ ~ ~ ~ ~ i ~ ~c d o~a,oNOO°~ ~.9~3., o'~"o.'~ ~y~ ~-o ~ ~'".a o ~o U.-.7zkc~r~'r~ Ufl:= ~ ~ ~~3~-0 3.~ ~.~ a»d>~
' 'O O N ...~ N 'G~, is N ~. i~ cv . ' ~Npa~ Wo~~~'O'~ ~~r c°cp.,o x .o ~ :~ ~ ~ d ~~ ~ ~.
~~ ~d ~ ~x ~, ~ ~, ~d o ~w ~o o .
W ~_;' o o,~ o~~ o.~ ~~ o'~
°~ ~N~ ~~ ~ °M~~Q~.l ~ .
odd ~ °.c~ c °.~a~~ ~4w.~ ~ o ~x~
~ ~NW ~ ~~~i ~U ~sU.=.~.~~ n..a o~~, ~s ~ x '~ o ~ p ~.~ ~a ~*~ '~ ~ H o ~ a~ ~
° ~~ b ~ °
'~ ~~-° ~.~'y:~°:~1~1 ~1Y7 ~ o~°~ ~ ° o oO ' - .
xQdH ~ ~.~ a ~x ~ ~ ~ ~ ~~.~ ~x~~~.~~

WO 01/79442 . PCT/USO1/11850 c x ,~ +. .~ '~ '~ > -o a. ~s '+, ~ o ~ ca o ~ ° o ~ ~ o ~ o ~ ,~ ~ o a~ o 'o ,~ i ~~,~~d~UC7~E~~Tr~a~~E~-~~Uwr~cn a~
o ~ ~ E . ~ ~ ,.°fl ,-~ .
.~ E Q O ~ bD v0 ,_, U U '~' cG N
'~ ~~ ~ ~3z~ ' O~V>'~p0\~VV~UO~
~ cG = Q, ~ U ~ N U ~ ~ 00 ~ pa d . c ".; ~ o '~ ~ ~ >, y ~c~ :~ av Q,.° v .' ~ ~ ~, GA v~
o~~ aD°''~o >° i v .~~
O U >, O O r., ~O ~> .~ O .~ p d ~O ~° W° W ~ ° o..~Ud~
a~ ~, bny o ~ o ~ a ~ ~
.~ °U' ~ a~ ~ .o w ~ _ ~ , a, ~ v~ ~ o v ~ °~..° ~ E .v>, .~ ' ~ ' ~ nn as a.~ .a s.' s~~U, ~.~
ccS V C~"C cc$ +~',. G' ~ .
cd ~ O .b cG ~ ~ ~ N ~ O +-' .u: C U U ,y,-" ~ ~"' ~ ~ iU.. ~O N O
d, 0 '~ ~N U ~ .~ U ~ U
-rNU~~ O~U~~.."NN' bD
y 'O O ~' ~ 'cc3 p ~ ,~ ~ ~~
x CL c~C ~ U d c~ ~ cct .O 5~,.~ U
~ ,...r Cf1 M d' ' ~ O ~' 33 3~
", ~ o Ur4~
~ b~ ~~~
a;
° ' ~Uvo~~ isz N is ~ ~ ~dQ ~~Cd~ o~~d o °~,';nQW
~ ~W ~~ ~.o~-c oct:~-o o.~ ~U'o ~ .n ~ d a~ . o ~ o ~ ~ d o c o ,.--.
~ U.n,_.,"~ ~:~ ~;.r~ o,~U~ od ~~
y' . ~~ ~~a ~~ ~~ d ~~x oPW.y ~.U
.-°'. od ~ ~ ~ o~ od.~~ od >,.~,;; ° .
:.o x ~ ~ ~ U ~o cc o .., o 'U~, ,~ C o U ° ~ ~ o W .
oa d o o.~,~ c ~ o'~ o.~ o"-~ o.~:°aov ~a~ .
~~ ~W ~E~ ~~ ~d ~~ ~d ~~a~., E°

w - U
p ~a ~ ~ ~ ~'-.~o ~
o x ~r Wa ~ ,~ ~ ~ 'a w ° '° . x ,~ ? '~° ~ ~°v ~ ~
~ " s. ~' 00 ~ -o ° :c ~o ~ ~, o °' ~ ;.~ . ~ ~ ~ a .° ~~ ~, o C7 -°Z O ~ o ~ ~ do '~, ~ ~~ o ° ~ ~ ~~ ° o,~ a.a N°M
~ a °°~ ~= ~~' ~ ° ~~~~ , .o~_ ~n~., ~ ~ o ~ ~ SOW
a~°~.,i°
°x m ~ =ro vQ.~~.;C7 ~ ,~ o ~.~Z"opo~,pao~~.° ~~ ~ y c ~o~~~~
~-° o~~ ~ ~ 0 3~
o ~ -~s c4 a ~ .~
U Y.. I.. ~ 'O tSS ~ 'Lr~' Y. °
o ~ ,.c ~ ~.'' °~ E-., v~ ep '~ GYa ' pa W p C - °o -o U ,~ ,.b ~ ~ ~ W O
~ e~n~ ~W° ~ '~ ox a;,~ ° °.o"a:°',~~ ~ oo.~~~~~
~~~~3a, N ~aS~f..w~4~p.~OcC ~ O~ .~~Y.
~~ ~ .~ ø,b z ~ ° O ~ N D p ~ a~-~ ~ ~ ~ ~ ~ ~ bOD~ O, ~:~ ~ ~ cC '~ ~~
V ~ c~G .~~O '~ p G
~ .G' .~ O +U.. 'G ~ ~ by E o .~ O ~~ O ~ N N p ~ O N ~ p .~ ~ ~ ~ ~ (~l C ~
~, N ,~ ~ O ~. ~
~p N cd O -O' ~ N -C ~p ~O p S.J °' ~ i-, cn U.. ~ ,U ~' ° '~"
~. .C c~ c~ 01 4~ ],~ U ~ .pu ~ CpC
x~ n...o UrnxE-~cn~N ~e 3~r''~'-~~t~ f~r.~ ~z.~~x ~Q.~ ~x,.'~'~ O.° U
~.~rmw ~ U -...
t.. 7-~. 'L7 N p ' pn p ~ U
Q Q. ~ ~1. c~C ~ O ~ ~r y ~~~~ N
aS F.' ..O aS by O ~ y ° wG4a y~,Uy" ~.
_cc3~p_OD~4..7..~S~~C~C~U .UuU~~°..t-~~~,~E'~~_~~N
~NrOn0~0~ :.0~~~~ O -Op"'pN~' N U ~ ~ ~ 'O
'~~+~.mU..'i.cNC>N~t~C~O.~~ Q.3~N..~'.~~,.t"~. ~.v~c~
U O ~.~. ~ ~ U ~ '~ V ~~ 'C1 ~ ~ G~ ° ~ ~ ~ ~ U F.' C"-.. ~ .
r-U.. +J bD ~ ~ ,.U. ~ ~ ~'.' ~ ~ +~ c~V . ~ ''~ ~ a"' ~ O CV -t'. O, O
~S ~ ~ , U ,+, .'a cC O O -fl O m .~-. N O .-~ '.i-. '+~
'C1 ~ 'O N O 'O .O ~ +~"r 3 ~ U .~ ~ 'h U .~~ i-. ~ U U ~ 7.~~ U
N~..~,~UN ~.C ~..;,C bD~~ TJ' 0:..'~.:' wi:~
o~ ~° N o . ~ ~ ~~ ~ ø,'~.b,a o on...cn a~.~ ~0000.~ U ~ °4,~~°°
r~~ a' rr O s.. +~ Cl. ~ +. n-W n .D L~ cC w.r a3 3 y.. O r.. U . U
V1 ' tn ~p d' ' ~
~ 'd' ~1 ~ ' ~W
O\
~D
U ~
Q>GIUv°~ U
Q ~a N o o ', °QW~ >;;~ °
v .
E ~ ~ ,c ~~ ~ ~ ~
w'~ ~ ~ ~ o ~~
~ ~~>,o~ °n~n o O o .N ~ o r~ o ~ .
Q O U ~ ~ ' N
C ~i'i W W O
+:~ . .
W C W W W ~O +~ - , ~bOQ ~ b~D~ a '~ W ' H u~
~ N ~bOQ ~p a . . d ~'a ' ~~ ~ ~~~'d U y-' M ~ . '~ O 'C U ~ 4.. ~ O -" C.' ~O ' .

~ .n-.
~ ~
c~c o?a, cn ~ o~i ~
GN ~dr:~ ~.~
oNo . a ~ _, .,. b ~~~Q ~ ;~y~~~ b~a w ~' xO~
~~
~o ~ _ ~ , , U U ~ ,_ ~ ~
~ .O ~ O
U ~ CMG N' '~
U ~ ~ .

.. ~
-v ~ y b~~'"~~ ~ ~"70WC1~Oyf~v~ ~O ~>,N
~2.0 > O G
U 'C~
cC

> p. ~ ~ .
'.~ C/~ W ~ pp,~ ~ ~ ,~ ~
~ ~ O c~ 4'' o ~ ~ v o -x ago ~

' ~~'' ~ ~ ~~ ? o ~ ~
a ~~w~
~ ' ~ ~ ~~ ~' .
~ ~
~~ o ~

, o, , w ~ -c o ' e~ ~
. '~
'~ ~
O ~ ~ ~
' ~

E.., H ~ j~ +' S N ,~ U
L~', Fi U a ~
M~ ~
O r ~
~
U
U ~

t-.~ 7,-r~-r ~,inO ~ 6 ~ p.~ l ~.,~ 0.~
,~'-'..O
~~
O.O, ~" '~ ~' .
S =
p ~~WN
~
O'O
~F

, 'a0 p ~V ~~"''Ca~E-~~~~~
~ ,00~~~~."
. ~
, C
~
~
.

'F ~V o o ~ ~~-;~~,o~ ~~
_.ou.; ~ ~ o ~o ~ i~.ca.

~-'~~.~UP.~~.~,..,,N ~.,y~~~~~o,~
~ ~
~
M

U ._-.. U ,~ ~ 4-n .~ cG U W ~ 'p ~ ~
U ~' J.. O
N 'C3 y ~ U
~ U ~ Fi c~
U G7 a!
~-' ~

.C .,.~" , ' O ~, O ~
h G L~
E"~ ~ ~ c~
C~ U a 3 E'' ~ ~ ~ ~
W
a h U ~.. .~ :-, C/~ V7 r >
-n f W N
N T7 C/~ E-U

W ~ d. ~

a U O U ~ ~ ~ ~ ~' '-a' ~ 'O .fl O

pp ~ ~ p V .~ ~ V V ,"n ~ .~

~ . V .S""
'"'" ~ ~ t~ Q 'cC ~ ~ ~
C

. O ..
~ '~ .~
U ~ c~ +~ ~
.

~ O a ~ ~ W > ~ O
U ~
~ O "' ~ ' t"

n ~ .' +.. ~.
.f~. by > ~ D~0 U (.'G ~cd"~ ~C+..J wn0 3 O , U O > "O~ 4-.O -~ Or."
O

O U

~ U E'~ ~.. b W ~ ~ +' O
O ~ O U ~ .fl N ~ ~

~ pp~

C ~ a ~ G ~ ~ O ~
O p O

~

, ~cU..bJJCy.O, ~
.~-r ~.U.NO '~"' ~p~p U
UO.h..,' N~~~~
n--~ U

~r R, ~ ~r-y, ~ ~,~ .~ -C v~ ~ is : ~n '~
a~ f.. 3 y ~ ~ ~
~
.
~
p 3~ ~ o ~,~ o d -'' ~ ~
Q
~ ~ ~
F
o~ ~

~ o ~ ~ ,-. u.
v .
.
o u a' ot~ o v ~

o o ~~~3a3 3 O~ M O~ O
O V't 00 O'~ d.
' 1 , ~1' O O~ V
O d' I~ O M ~ ~. a1 ~O

O O O O pp ~ N .-, rt d' O O O O ~ ' ' ~ ~ V1 ~
L~I~I(~'~I~ ~O ~ -zxxz33xm~.3 ~

. , a .
F .

o ~r-,~
.

~
. a N U O
7.. '~ N ~

d~ a. Wn a~~o aW -' ~ N O
F ~ l~ O ~ ~

O . ~ (~ ~

N N W .r; G~ ~ C/~ x 'nU
o N O ~, o .
.~r ~ t3 bQ - .O ~ H ~

.
~i''~ U.~ ~ ~ ~.

a aQ~a~

o ~ .~
O z..
~U~
~.r. ~ ~ U
. m . x:, ~.. ,n .
a~
Ll ~
U
C ~ ~ ~
'~ ... by o a ~ x.~3z ~~ ~ ~~ ~~
p ~~ o ~~~~z o o *; o a. ~ ~~ r~ ~
.~
-d o o '~ p ~ ~ o °°
~~ U ..D ~ x '~" M , N '~' +~ N ~. ~ of ~
c~ ~ ~'.~ x ~1 ~ C7 ',~
c~ ~, O a vi a ..0~ ~ ~~ ~.~x~°
v~ ..... 'O N " . ,.fl p, t-. ~, ~, c~ > ,~
rn ~ ~ ~ ~ ' U ~ ~~ tC U 'C3 U ..'. QJ .n-, U U ~ O
N Y '..' ~ c_n O ~+~. U O ~ O ~ ~ N ~ ,.G ~ U m .p ~.c; ~ ~-c ~ ~ i ~ ~ °~ ~~U OU U
~~w, a,'~3+~
oi~~~~~T'o.c~~cs~~~c~a.~~,$b~~"c~a.~.~c ' bn o ~, a~ o a > a~
a~ +U.. O .~ ~ .r~". .t_C N N N pp O ~'~'~' ~. ,.U~ ..N. ~ b .O L: ø.~
U~atS'..'O~U~Y~' ~~O~~OC,UO .~
~ ~N ~ ~ ~ ~ ~ ~ N ~ ~ 00 N .r'~.' ~Q..b O ~ ~ ~ .O .
~~' i~ . VI
~ a"0-. . "err" r.-r~1 bA a1 Y~ G1, +~. 1u '.d tl-n ~ .fl . ~ ~ cO U (C CG
.'~~
M
N ~
~ ~n ~N ' .
~.~a , _ø._;_~~
CC ~a U n N G,' ~ ~

~o~~ '~
°U~ , ~'Ga _~ .on ~
E ~ i o o'b N
.~caC~v~z~~
l~ W ~ 'N ~ ,,~b.o o ~ o ~ t~ ~ ~t7 ~r:., ~O ~ ~~ ,> .Y ~~ oo ~ v c ~-°..~~ vGq°~ ~ i ~ ~~,~~~v ~'~~+. o ~ o; ~a ~. =' i .~ ~ W c~ > :~ .~ x a, o ~ N ~n o ~.;a Z c~i ~o ~
~, ~ U
~,~.o ~U ~ ~ ~~.~~My ~ a ~.o.~ o a ~ c~ a~ a .~ ~ ~ ~ ~~ ~ .> ~ .~ W G7 ~ ~ p. r ~'~ ~~"~ on> ~ ~b a °, C~~C ~ c~~"C ~N ~ ~ rU-1 r~i7 ~ ~ y 'U
o c 3.~U o.o~oo>,~ ~ o.~ ~ o ~ ~ o~~ . o >,o ~~ ~ bpp0 O c~c ~ ~ ~ ~ ~ ? ~O O ~ ~ ~~ ~ v ~ ~Q.. ~ y U N
C~ ~.~aa~UN.o ~Pap~~ ~ ° °w' ~ ~~~.~a.~ ~W n V O O '~ ~ ° ~ O .C V' o f., .'~~J' O O N c~ N ~n ~ O O ~ ~ , O 'z3 i-~ ~ v~
~ Q.~ ~ o~n~ ~~~c ~~3 ~~ o~
caoO~om~°iC~ o.'~a' OC~.~O~-C ~°~-p'p ~~~ ccl~
o.o o ~.c~,~ o ~ o ~~~ ~t° °.~,o,o-o'~ ~~
d- ~ bpJ,..... O ~,~0- ~r..~~N~p 'C ~ +-~ ~' .~ '~' _~ ~° ~ ~'..*'~ N H ~ o..' ~ ~ c'. O C .~ .~ b N ~ -O!", ~ tOn N
cCt VbD~~~a)~~°ObO~~Oy'X~j~~~~~N~~~+~...~N
E"~-' ~ ',~ ~ V ~ ~ ~V '~ ~ ~ V ~ ~ O O O ~ ~ ~. ~ ~ ~ ~ ~ ° ~l X
~O
cC cG ~ cct w ~ f3. ~ of ~+-~ t1. at ~1. v~ . 'CS s. +~ cC vW -, N
N
O~ , a1 N , N v'~
a ~. ra a~
~, U V
y. ~ L".~, ..O O
V V
a.. ~ O
~ ~ dOD r' r~
~ ~ C
O
z w ~ °Q~~w o°n,o~n~y ~z ~~aza U
C
ccf Y.
U v E~ ~; U U
'G U bU ~
~"» C ....
O cd ~ O
v~ U~U
a~
ou oo .
o ' N '~c p ~... ~ .+, ~ _ o t~ p ,-~ ~n a o C~
O -fl b ~ ~ cct ~ ~ :x O U O U ~ ~' ~" ~ ° cn N ~ c~C ~ ~ ~ M O .~
p ~ ~O
~ .i~ .G. O E" O cG U U ~ C d' ' ° ~ Q 'U o > ° ~ y cn +~
.,r : d ~.~ ~ w ~ ,--. -o ~. ,.o ~. ~ ~, ,.o w w 6, ~
'~ ~ ~bA v c~.~a ~ ~ a H Q ~ V 'gyp w ~. ccs ~ ~ ~ ~ ~ ~ ~ .~ .-. ~ ~ M ~ ~ ~
G.i ~ _Nx ~t i o ~ ~ ~ >U-~ ~ ° ~ aE"
~~ ~~~H ~~~~a~., ~Z
C7. ~ N vy0 ,y ~,~ C cn .c '° t~, y., Q, N ~ ~r~n ,~ ~ ~ .O ..>.. ~ ..y ~ ~'~~' 'k ~v .3 C U W~~" ~ ~+. N
~.-.~~n.~~ m~s~.~ a~. U.~ U~ a~~sa~ Q~ goo ~~~°~ocn~ "d ~.fl ~ a.~'a~ o ~ ~ ~ ~ ° ~ iU ~~'~"' o."o~ j wa ono o x.~a: ~ ~ ~-o~ ~
N o ~,~°,w~ i~ °_' n.'.G~r~.U ca w~c7C40~U ~:o,~'U o~a;v~W t~ U-o a~Ucda,zr~ ~U~wW
U U _ N C .a:~ N U
"d ~U U ~U U ~° U O U ~ i~.~ , v~ , f.,' "'~ ~ L O ~' 't~ .C ca ..d b~0 cOn , ° , m cC N ~ .+~ .C
~ W ~ G cC p U V7 > ~, ~ Y ~~3 --. ~ ~ bD d' c~ a..~
w O N
..C°bO~D'O1-y.w~~N ~UU
bA 4-n 's., G ø, U cn ~ ~ ~ cn c~
~~ t~" p 7t fn ~ . b O p ~N
~~NU~N;flUG.I~~~,~ ~X.~
~3 ~ w ~. .n ~ > . o ~, ~ ~ ~, a.
t-aTU~".I"U°~FiUN°i..' ~N
O~y~auC~~'_.~.'.~~U ~~L:' ..v~. ~ U > ~ N O_ ' vi ~U y +, ~ >> ~~-.
cn~'>° ~.0~~°'C7yU, N
G~ tOG ..fl ,j" U .u~ ~ ~ U Ct. ~ c~ O "'~ a O
~ N
a1 O
r - ~

f\ ~ ~ ~
N cal ~
N ~M_ ~; oa a 3 - ' ' U
~ o -oz ono boo °
',~, ~ .., ~ '+, ~ M U
~ ~ ~ O
~G. ~ ~Q ~' ~ .
~ it ~ O
b N > ' ~ N x ~O
n"~ . ~ a ~ M
.9 . O b . 'p O ~O ~
v~ M ~ v, M Pa U ,.U. G~ V U a Q _ 'J, ~ ° Q1 ~~p 'J~~ ~ ~ ~ ~ '~
~~'°.aaN ~ >~J'~NQ~ .
.~ ~a~~ .~ ~ac~~ ~ . _ a c.

E

.

x ~ ~ ~
~' ' ~

d.~ ~a o ~~~'~~~
o ~ ~ ~ s'.. O ~ ~ b O O ~3 ~1. O c St +J t r~

_ _ ~~~sa~
E-~ air cn f~ d La v~

~

Q
d .~~~ ~~ O N S.' V ~ ' p W-.., ~ ~ ~
' ~
, ~ N

, ,-, G', U ~ '+~ 0.;
' O .~
p ~
fl vo ~
..
nU ~ ~o ~ ~
' ~
~ ~~p ~
a N

, a :
, ~
.
, ~
~C7 ~d-c a~
;o ~

~ ' . ~.
~s o~
ba~
~

'~ ~
s~o ~W Vo''-' ~ ' ~~ '"'.~.~

.
'Uy." N O > o a~ .+; p G

~

.;, V . ~ cd .~ t~ v1 U
O C/~ ~ ~ ,cd G
y O ~n O b O ~ i. bQ~ +~ .~ .~ ~
O

' ~ ;.O ~ ''~"i ,~ C 00 N b.0 ~ O N' f +r' d - .~ ~~~~~
~~~~~~ ~~o.~~~~~~~,3 ~~1~~ ~ o ~o, ua ~
~-o ~ .~ ~~ ;v~
._ ~~~~
~~
x ~' ~
~

~, _ y O
p U ~
~ N
o .~~
_~x 'C7 ~ yn ~

~. -~
"O tY .
' ~, t~ U
c~N
O~v ~ N c~O.
N V G1.
.k ~ ccf .
O .~ ''" ~ ~O U .f.~ ~ V ,n ~
O
Cue p r~ U N ,.L'. .~.n U ,~ O, ., O .~i ~ ,.
~~ ~xU~zH ~ , ~~ ~ o 00 ~ ~

~

. ,.
~,.
.
a -j R O
O 'CS

sw .gin.. .~ O
-,.~ a ~ ~

pa ~ Ga ~ P.i ccs pa at >
V vi ~ "C
d ~

_ :o ~
a~ ~ o .~ ~ c ~ ~i '-'. t~p.~ ~ v '~'' ~ ~ b -fl ' Wn ',,., i-~ N Y r~ Q CC d] 4-..
br!D ~ ~ .~i N ~ ~ ~ .a U "~ ~ ~ +~ Vl ~ b ~ O ~ c~ S.,' O ,.d O U
U at '~ f~r ,~, ~r N U~ ~cC O ~p..~ , N
~~ U

~, ~ , '~ ~" .
~0 ~~
~
ba t.*' F".~p. ~~
N , ~~
O~W~,C ~ Q, '~ ~ ~' ~ p O al '..3'W~c U ~ N +~ CC t7, ~ aS
~ ~
4-~ N
' -"

.. ~ .~ O ,.
p ~ as ~
+ ~
C~ ~ ~ w3 .n '~ Y O d O ~ +; Y ro .Q ~ U_ U ~~. a~
~ ~
~
~
U

U O~'~.YN 'U~N
~NG~~.~ cc! ..Gl.
~0 CC~ +'_S
NN.~
'p..~.~
~Y
i ~

~. ~~~~ O N.U., c~ O t3.~~',~J,2$W~~
~N U~O~ O Uv~"O
~.O ' s.
S
-'b .
.~ ~ .G " .
~ ~ .f O~.C ~ cd a '~ Q 0 N
~% '~ ~ ~ U '~ ~ ,~ ~
m ~ G.. Q.' ~
Y ~ jY
'~ y ~ r..
:

: t , N ~ . _ al U ~ Y 'L3 O cci ., .' ~ ' _ ~ CG .S"" ~~' U cC3 O ~ '>' ~ ~ ~ a' ~ U y.,' +
'' . U i tUn "O
'~
" ~ N U

~.. ~ 4w _ ~', N ~ . ~ r S"-.. TJ O .
t-a .. U +.w-. U ~ d .. Q7 ~
a...
O O S-.
~ S~,~ U U cV -..~"
U ~ at d at .~.. O
TS

M

_ ~ N

' 3 ~

. ~ ' o Q ~ O ,.-, o a. ;~ U ..o . b Z r~ ~ ~n ~ , ~ V .
~ ~
'y CCS .-~~
I~ ~
t W
.U W ~ C
W

cc ~
~ . ~ bn c~ 7 ~1 ~r '+:~ ~
U1 F'. G '+J

, ' . ~,..ab.~
~C7~~ p '~'~ ~ ~

N - tUn O ~ ;.O O ~-, ~ ~ . ~ ~ 'f-.' ~O
~ v~ '~ ~ ~ ' by .
C ~ .
: ~

. o W . OO. . +
~" O f~, O . ~
Vy..a O : f.~~" O n -~.

. ~

.+~ O G T-~ ~ _ yn O ' ~ ~ ~ d O ~ ~ ~ , ~

O
N

N '~ f-~ ~ ,~ ~ o ,_? ;~ b C7 A ~ o U o A

' ~ a'' ', cy ~ ~ . ., Q :O ~n G W t~ , C
~ ' '~ U o~ tea; Q ~U ~ d ..
Q .
ate ~ ~.

a .~r~v~ ~w ~x~, ..

x OU H

o ~ i ~

o N ~ o F." ~ ~
. U

tU., ~

pr C/~

_ _ ~', W n ~ .,*'~
O b0 > aS
'~
+' ~ .v .~
~ ~
c~
,.~
U ~
~ .
v 8 W ~
u _ ~,.
, .~ ~
~ ~
o ~
a~
b L1 ~:
cd 'H' ~
U
4~ ' W ~' O N
C 'in O
~

O H
,.
.
:~~0~0 ~Gc~CU~VO~~0 ~

~ f:. 0 c ' ''' ?
V
G s... X U '~ . ~
~
:
' m y ~ o U . ~a,a ~ ~~ ~
~ o ~,~ ~ ~~ .
J c a ., ..
v r+~,y~..o~ ~
~ Ua~
ovW~.~a~
~~~~y ~.b~a~ ~.~~

_ ~.
.
~ '~' v:
~'~.' U O
~ ' ,~
. ~
N
~C ~
cUa b4 Q a O p ~ ~
"C7 ~
U

, O
c c C
y~c.
, ~fnaiCl~
~~13 00""O
p ~ W

~
, .
n ~
~
~O

t~+JN 'C
NG~NN, ~,N~'~
UOO~~>Gtl.~~O~
' ~
U
, te .. Cf U
b~p~
~
U '....
O G
U
CSC
~ ,a tp CG
U
~ ~
~ ~
N ~
~

~ ~
~
~ o ~ ~
.
~
3 ~
~'~
o j i '~

ca ~, o x ~
~
.
~
~ O
.o a~ ~
~
1~' ~ , x '-' '*' ~ +.. M
, , .
....
~
n W
., a~ o ~ cc~
~ O
U ~
o o ~
~ ~a s. ~, ~ ~
p "
N ~ z~
N ~."' y U
~
U', ~ ~
,' hU
U
~ .fl N ~
O ' U

, .
, ~
~

~ O ~ ~ c~
~ ~
~ ci ~ s., ~ ~
Wn ~~ f1 ~
~
~ .~
~ V
'a ~
~
O

C.~ Cl~U
U.~V3 f3.
13..~a.U
tn4~~0U
V~~~O~NC

N r. o E
.

U
7 W ~ ~O
y ~, ~ ' O ~' .~
.

0 ~ ~" ~ ~ ~
.~ ~
N ~
,~
~

0 , N N N . tC
~ +...d~U
UC1~>H~ a O ~, cOG 4.r O

-0~.,~'OY~ _~
~ ~r~nU
c~
U

~ .o _ c~a -~, ' ~ , ~

~, +
. bn ... .~ U
~U , ~ U
~

i~ P, . U~~~
a30 3 , ~ ' ~ i ~
~ ~ ~
w ~ S
, ~. , b ~ Yr s. w , O
n O
O

S." .-~ N N
.4"'.. O
L: "~' tn > ~ .
n ' C ay ~t +
~ ~ ~ ~UU
~c ~ U o ~ ~
a c .
c .

M

~ ~ .

, O "' 4-~
w a3 _ O y ~ U
v0 N ~
l~
. ~
a1 ~
~

.~, U c n U
C
b0 +' O~ U
~ ~
ck1 . ~
t,~
O ~
N
~

~
a~ t~ p N
~ : ~, Xp= m W ~
N
~
l1 y ~
O LL
~
~
U

' p ~ ~ p ,-,-~ p -..
O ., 00 G ~
N U
~-.
.
U O~

_ ~--~MOI~
T~~ M N
~ a.~
C ' p ~~M.~
~ N

O
d ' ~
' W

, ~
(1]
v 1 ._.
~ U
4-.n ~
n ~
N -O
~ ~
~

, oU ~A~d ~ ~~
o ~
~ ~
~
x~~.c ~ ~
~o . d d w~~
c..~' '.~~'U~vW_ o.U
~.._H
~

d . U

~ . U
.. ~=., v~

xQ

o N

of ~ a U
O N
O

x v U:9~ o a .

~d ~ ~ o y U

_~

O
Q, j~
N

4-. N .(C O M
~

~O U v1 ~ v1 .b ~ ' h O
,n ~ ~
~ ~ ~ 4..

M _N . .
O ~ ~ x . ,-;
-o .~ ~ D
F. ~ x ~ ~
~
~

S1. i.. at O M O ~ M ~ '~ t cd cC . y ' ~ N O N O
~ O
' > .~
O +J
~ M ~
' c U ' ~ ~ V '~
' G ~ ~ ~ O .*~ GL' N
~

d ~''~ ~ w ~A"
.~.

a ~0,. ~ M
U fn ~ ~ C ccf ~ o ~" P-i a O
~ ~ ~ O ~ O
O~

. LL ~' k ~', ~ U V
., ,' O ~O a v O N 7 U 'Z3 ~' ~ S'.
cG

~ ~; N ~ ~ ~, y ' ~ .fl ~ '~
~ C ~O ~
N ,~ a , N T3 ~ c,.,,' -, ,~"" 4. c~ ,-, O .O C) -, .,r ,-~ o d ~ bA
U ~ ~ C ~
O, ~
. W ~

U a . ~
U u~ ~ ~
~

.zyE" ~~>~Z~:.~ ~~ ~, :ou.~~,~' ~' v ~
~
b ~~ ~~~ ~a ~ X ~ ~ ~ ~N
. 3 ~ '-..

~ ~f3.r-7~ 0. ~~N,~~"~C~CNO
N ,~S", , ~', ~

" ~ a,zx ~U~w~
'' ~U~
.
.~;~~U
O~a;~W
=
-o U

O
O

~r ~

N U

p"
X

N N

L", .
'~~' O
~ Lr ~ O
N

bD
U

N a1 N

pp N ~v~., O~

O v'~ . . .
~ v~

~O

~3 M . .

'a ~

U

~, ~

O ~ , . .

U F.' "d U

O ~
C

V ~
_ n .

~ a ~

WO 01/79442 ' PCT/USO1/11850 E
°~,' c~ ~ ~ d " +~ -o O d ,~, t~C ~ U
*~'' C ~, O , cci ~ Wn Cc3 ~ .G FUr~ iv N
O yt~-..'n0 4-~v~~N~~~-. ~~'~O~r~,~U
w~~~ O ~.~ ~ ~~~~ ~.,tp O ~C/~V7 b0 o ~ oU~Q ~ ~'~'~~ ~°~. xGG1 c ~a~o.~ v on ~ ''" ' ~ on ~ .~ '~s ~ a. d ~ C7 N o '°
w v~ o ~n ~~z.~ oo~n~~ c ~°o ~~ ~ o i a~~a: xr~.';y-o~cox~~°doon.~~~~ ~o°v~,Z.~f~
~ca~c._~.Qo . ~ . o ~ v~ ~~_o"'°vW~°
4Z." Z ° .o '~° ~ ~ ~ W ~ ~ ~ ~ ~ ~ ~ W ~ C7 '~ ~ ~ ~ c°v .... a. ~
~a o °o Q '~ ~ ~ ~ :N ~ ~ .a ~ ~~ c° '~ o ~ ~: pa ~o N
y.. ~ yn ~ m ~ .C ~ ,.. 00 ~ o ~ E ~ ~ Wn v~ c ~ ~ M o'"u ~ .~ N v~ d vi d G. C~ p. Gq v, a~
+: y~ y ~ y a, o ~+~
Y o ~ '~ a c . :~ °' ~'~'a.~~ >, ° ° ° ~ ~.~ T~ o .-, a~
0 0 wC ~ w > '~ ~ Q., o U
~O ,~ ~ cct , cC ~O ~ .~ Wo U .O
+~ O C
i.n CG ~ Vi O CCS ran O f~.Ø. . ~, O' ~' Q) ~ O ~ N .
~07-.O~ ~pVU~~LLUC~..v~
N N '~ N ~r ~ V ~ V G.' zoz~ z~~.~a~.~~~
000 N o0 . v'1 V1 M 01 . ~ 01 Q ~ 01 G3r~ a Q
zap a -b o ~' ~o o ~~ . .
0 0 " . .~ , o °c c ~ ~l z ° E Z °
°E~~ . °
o ~ ~ , i o~~T
d,s~°Z o o :flZ o o +~ . ~ .;n , o : +~
z~a WO 01/79442 . PCT/USO1/11850 c z~ ..
U

a 0 G o U ~ ' '+~r a..

U

r~ ~
_ U

~ ~ O

OwU ~ U

cf., U ' ~ O - M
O ~
~

O ~ G' _ .~ ~ . V1 V1 N ~ h v1 O ~ , ~ '~ by ~7 .~ ..C ~ ~n ~ i . .-.X ~ ~~ ~t X y-;
cn. a ~ ~ ~ O . vi N ,x v~
~ ~ \O ct M O s. N f 7 O N V
c ' , C . M p 'V O ~ ~ N ~ O _O
N y- . ~ ~ O.M
' p ~ a 'U ~.~ ~
> .p N

p ~ t.~ ~ ~T.~ N N ~ aS ~ 4..Wr ~ p N ~ O
~ ~ ~' ~ ca N
~

,+~,,~O ~ ~'' f~. 4Tr ~ U .b U Q" l ~

'''' 01 '~ +~ N U M ~ . by cOC
w O c~ ~ O bD'b ~ .~, O

U a ~ Q\ C: ~ U M d' '~ i'' t-i i 01 O .!', L1 O 01 O "
U ~ ~
~
p C ttS Q ,~ ~ ~ .~ 4~ CC ~ ~ ~ bf1 , , .~ o cC ,.
. U'E~ '~"
..
O ~ ~y ~, -BOO ~~~a~~ O~ ' a,c~
~ U ~_ ~' a U

~ ~ 'O ~.~.-~,~ .n-. bA >' ~ ~~ > O O O , '~ G'. (~1 , ~ > ,., U bp L: 'N ~ ~ ~
O r.. ~
~ ~

O ~ ~
U O ~ ~ ~ .~ ~ ~
O ~
~

.O ~ EA"'-' U ~ ca ~ ~ O. ~ ~ O ~ .U ~ p L~' y Q ~ U ~ ~ ~ ~ ics ~ ~ bU0 ' ~ .p ~
'" ~ p .

.,=, ~ ~ ~--~ a~ ,O O
' Z . ~, .-. . ~ o .
~ ~ ~ ..o ~' ~
~ ' ~

~~, 3 ~ ~ ~~N ~ '~~aa ~ ~ ~.~
~.~~~a.~w o b ~

0 x ~ ~ N
> ~ ~
~ a ~

~ ,..'' ~ V X G ~
~' U cC G. ~, W ~ ~ x O
0 ~w z U z ;~

O a ~ ~U w ~ O ~
~ Ub r aa b~
a, r :~ . o ; . ~ .~

+V.. fn ,yn N n N . O W ~ N

N a3 j., t-. U .O TJ ~
ce ~ c~

... Q ~1" 5" ~ ~ , ~S. ~ i "

U .O ~t...
+' L
, N
~ .fl N
.

.' ~ > ~ c~ ~
+N.. N ~ ~ O 4.-n .t ~ .~., ~ ~n Of~.I..NO
. CObp~~. ~
U ~
O
U

Q' _ UU
O *~'' O ~..L~~ G~,~
.~'-n ' T ~ ~ ~ . .O ~ 'CS ~ ~ N ~ ~
N :r ~ O p by N ~

~ by b G. . U U . ~O b ~ ,0 i .~ ~ ~ ~ ~ ~'~ U y U

O ~'.'~ ~~~~~'~~ O'~~.+,cG

.- .~,-l~. ~~pUOp".C~O~S
'VyU,c~

P~ ~ Qx7 U ,. G.a ~ O i.U.~ ~ 4~
U N N
~ U c~G

O

V7 M_ . O

O ~O V>
~1' O

M _ N O
M O

d . . ~ a\

N M p~ r.
~

OOH . N . OOO
b O
O

' z3 a J~
zx~

, w y ~ p ~ O a ~ O C
'~

~t ~ ~ O ~ O ~ ~ O
. C
U ~ ~ ~
~

.
C ~ ' Ci V ~ O N O
G. . c~ ~ U

o '~

W
~

~. w .n ~ U .c ~ a.. y ~ c fl. o ~ ~ ~ ~

' ~ fix" 'ai a~ .
.~ a~ ~ U ~

U y ' ~
~ O a ' U

O O :~
O 'O ~ ~ ~
~ ' ~ ~

~ ~ ~, f1 ~ ~
~ _N O O ~
r' ~
~

~ .D f3. Q. ~ p . O ;p by p ~ n0 p p .~ G1 p . j N
.

. ~ ~ . O ~
~n i O
'~ O
'~

E-~ N , ~ v W ~ ~, n 5D '~
w ~aaa U 7, a l 'r ~aQ
' gy ~ ~
~

~ , .
, .J
p., C~
L
bAc G

o ~. ~
N ~' U

. _ ~ ~ p U N v1 v > N U
N

bO~O N ~,,~J

~ C7 U~
U~1~~

O . ~ ~' ~ U b ~ O
f"
> -.
d ,. ~ O O ~ O ~ O O U.
~ ~ ~
~

C. ~" ~ G' C. vwf''~" 'iJ O
N N
o o ~ ~~

~ ~s:o_ E
~ ~,o ~;
d o -a ~ ~
o ~ ~ o '~ v a ~ U ~ ~~ ~ ~ ~ "~ ~ ~a ~ a1 ~ a' a~
a~

,~ c~ C!~ ~ ~ t-U" cct .C
~ N 4'' O >,'O v1 . ~ ~O.a ai .O O
_ UU jON~~~-, b.=.

,~ 001 O ~? O ~Cn ~ ~~ ~V p N

0 ~ U vwn ~ ~ ~
W.~. o0 O .U ~ ~ .~ ~ ~~ ~ S"... ~ U
c~C ~O '~j cC O N
'.~.. ~

OO ~ ~OOOW1" 00'-~a~..~
~ ~
~ ~ c~ N p U
' U

i-~ ~ .. ,-o c: N -n o ~ +
~ U ~ > ...
~ ~ ~'! o V ~
~ O

>, , ~.
y~Ua ~,~.~~at~ _ - .
:''~a~.UC.~ppo ~~g ~o~~ :~~~~~~~~
:~ M
oz . , ~ o. ~
~ ~ W ~ ~ ' O ~a a, N m ~ > ~zN ~~ ~.~ ~Nc~~

-~ ~ ,, o ~ ~ ~ ~ ~ ~ b.

b U U N ~ . ~ ~ dD .y N 'C
~' ~ ~ ,F'' + ~ Q.' b0 ~ O .C Y ~ O U
.u . ~, cn L~
~

N ~ O ~ .C -G O ~ ~ ~
O . ~U. ,.C ~ ~ .v ~

p .

~ ~~~~o U' ~~
~

c a~ v ~ O
a ~~
C ~ N O
w ~ t7 .~ O ~ y +~ . O W c~
~_ ~
k Q. w .'~ '~ ~ U p. l~ ' ~ N .~'' N a 'in L1. WN N O ~ ~ ~ t N

_ *~' p, ~ ,~ ~
~ ~ ~ ~ N
~ ... ~
~ "C

,r . --N'n v .
O U
.
N

~ N U ~ ~,' 4-r .U 4r O N
+U.u y .-, f-.. .
~i ' N
4-n -' O

:~ ~ CC ~ ~ 'O ,~
M ~ ia3. w '~'r O U O c~
~ U a~~~U~
r a~ ~
U ' ~.cz. ~ a ~ v; s~~..
~c~ ~~ ~
O ~ U..O~
O .O
O

O. ,-,~ N(V4s . UN w "O ~
, C.
~

a; ~~~ ~ ~ .
v o~ .
~ (~-~~ ~ ~ ~ ~
~'~ > 3 v .
c ~. . ' a, oo M

.

c~ U

, ,,, T f3.

N ~ O ~"
O O
~

CO pp +, U
~ d p ~ ~ ~"' c~
~
~

b O

~ ~ n ~ <t ri..~
~ N .~' C O .C"r +.c .~
~

, 3 cC
zx3~

c~.-N

-, ~

'd ~ N~ .~
-r O O y b O
O p . ,~ ~ ~ ~ N
' U ~ p ~
p ~. ~ a O

a o ...~w~ ;~~~

O ~ p N
O ~ O O , O

t. :.O .
N ~ ~ U U ~ ~T-~ ' U O O C/~ p ~

~ ~ ,.~ w. i N ~
y Q., ~ ~~ w .

.'Jr' C/~ V
~ ~ ~ 7~' p~

o. , U
C ;a L > s~-. .N
ci ~U ~U ~~~ .
~~C7 0 ~
a~~~
p.~.~~ ~ ~G1 0.
'~. _o o °' x ~ a.~ c ~ ~ ~.~. U ~ .~ O yes., pra~a~~xE'-' . N
*' ~ N
T o ~ . ay ~ U .°c ,~ o ~ ~1' ° "O ~1' ~ U7 O o0 p O ~ ,~ 3 W ,y~ ~ ~!
'*.'ONU G Np_~~'3~0 ~' ''Y,~ .~'U N.
~ '~ >, r.U. O .~ "'~ x pp +' N C/7 U M y p U cC ~ ~ ~ .~O~. a l~
N t~.U.r ~ T ~ M ~ ~ O ~ ~ ' ~ ~ ~ \/ ~ O ~ O ~ U
O 1~ ~ a~ a~ ~ ~..'' '~ by . ~ -d O
U ~ ~ M O ~ ~ ~ M '~ U ~ ~ V ~ ?, ~ ~ M ~N U U
E-~ o ~ ~ bn'o ~ 3 0, '~ o ~ ~ O ~ ,a +3 0 ~ $ '~' ~ ~ rry a~ ~ ~C .~ .~ ~ °' 3 n».~ o ~ ~ ~ .~ .o ~ o a~ o ~
~ v ° ~ ~ ~°n 'n ° o o.~ ~~b~ ~.~ ~~~,;o~~~Z~r., N o~U
~~.~0>GmN~p~b~AV~°~'~~y~.,c~C~E~'~'00~Y>,~.:ON
ONN~.f~r~U~'~»E"~~N~~ON~»~~~-.-,r'-~!U'00~ i .o ~O 3~ ~~ ~c ~ ~C a~ ~ ~ ~;~ ~d- ~.fl "'°
a, c_C ~.-yN O~ 00 a sa,d ~ ° ~vW ~~8 01~~b ° ~~Z°~U ~ ~:~~
~.
° o o ~ ,o y ~ , ~ ~ ~ ~.~ ~ ~ ~;> ~,H
V U
G UQ U ~ U + U rUn y °
U '+u p, ~ O .L ~ ~ U ~ '~. -p +~
~ . ~. .,:. ~
~T N O U ~ ~O O U ~ N
s'..' f..' .-.. ~ .C p ~ ~
~ U ~~ ~ cUG C p ~U U
.9 ~ H .? ~ ~
~ o 'd ~~ 'n x~~a,~.~°.~~ao ~ N
O ~ ~ ' O d. ~
N~
v7 a, ~ ~
p ~ O O
O O ~
O O N N
~p v1 « M
G~ P, G~ ~
r ~"
o ~ , y c~ ~ y, .
~. H N .o O O V O
U N E"~ O ~', ~ ~ b 0.~ M
~a~~, . °
° x ~ ~ ~.
U
x ~ U ~ .~.. (3r G' N L~. ~
~' y v~
O U
C~c~x~U VU
°
w +~ N cn n -o o ~ "' .~' o ccs ~ ~ c°n ° ~ o °° N "..' ~ U ~ ~,., ~ w ~ ~ v ~ d a, ~ ~ ~, o O
-~ E~ ~ °' . ~ ~.
q x .+, ~, .d " ~, ..' °~v~ ~HhjH ~
"d Ts ~ ~ _ ~ ~ ~ o c~ ~s .o ~ :-. ~ ~s c~ o ° '~-r ~ E-~ ~ ~ c~ ~ ~ ~
> ~ 15 ~ ~ 'U ~ ~ ~O .~C'- .~ ~ ~"' U .~ a3 N ° ~ M ~ m ~ N U .~
~ ~ +' ;N ~ ~ ~ W ~U ~ ° U ~ ca U ~ U .fl V1 N bO..W'r N
~..OCdl3.UNO.~~~~~n~~~OUcnC~1+, .~.~'~C~U~N tYG+~Ov ca ~ ''0.~~0.~UUso.Nrc~ 0~7 ~r~P~.,~ w i~ v.~ ~UO.~ ~a~ v c~a~
+~
a ~ ~ ~, ~ ~
0 4-. ~ ~ ~ o _ ~ ~ ~
a~"7 > vi ~' ;b ,~U, '~ ,~ by ~ U U
U U U ~ N ~L U cti U k ,b~A ~~ ~, yf"..' tU..' N ~U .~ U N E~ 'O 'ate.. ~ O i ~ ~ .~
F' O
~cUC~;+.~~~~ ~U~CJo r.U.' bD N U ~ ~ 'C3 ° Q U > f~~ U ~ ~ ~ U , ~C:i.~.~viUUNr'..' ~~U~'L:
~~~E.,y~O~~ .~~~~
a~ ~ .~ ~ U t3. ~ b4 ° ' - N ~ ~
H G.' N U 4a N v~ L' ~' L~ U ~, O c. .~

.5.." t~., U N x' O N ~ y Q~., ~, ~ ~
°~ >.~-v E-~ ~.~ °
M
N
O .
O
°1 ~ ~ N '~
.-, ,--.
O ~ r-.
O O d' c~ cn ~n M N N cn r P-~ I R. ~ a a a O .
zx aaa~
;"
U ~+~
O
C
~N
c7 N U N
'~ ~ .
C~
. . ~ ~ ~ .
N
C U W
.U
b0.~ A; ,~, .
';~ C > . N N ~
~.a~°n~, ~Q~,U
N ~ H H ~H x 0 o c o Y
N O ~ N C V U N C
v 'N x ~ 'N ~ ~ aS N
C O ~.' ~ .T. L_"~" a' ~ d. Ccr~' Q.
,~.~.. vi G' c~C C ~ U C cOC '~-."
U~cC1NL_ccSLLJ°T~c~C
~~Q..~~Q.~Y~~~., y L
f. v~ s. U .-. rn U O
L1 O 7-~. ~ o L ~> ~N
df~E-'C7~lE-~7;~W nE~f~,'r~ U
p ' C~t -~ ~, N
°r '~ W ~ °' U en U °~ o ~, ~ ° ~ o ~ ~
v ~ a c N -~p .p ~ ~ ~~ o ~ a. ~ ° ~3 CC c~ U . ~ M C 3 i-U. U ~ ~ x ~ ~ ~ ~ M .~, ~
>>, ~'~,~s~, ~~o>, ~ N~TdW
W o ° ~ ~ ''~''" tx., ° H o o ~ '.° pL., Wai '~ ~ 'r o ~ ~ .~ dy .
~~ ~~i n,~~x~-o ~ T ~ vN W,~,r~n ~'~ ~ ~-° o ~,.°x, ova p . L ~ a~ M ~ ..o a " Wt ~ a~ >, p o ~r ' ., o o .x ~ a' C ~ ~ d '-' ~ a~ at ~j ~ v-, c~ r.. ..fl v ca ~ ~ ~ i~
~ ;~ :'? ' .c v~ ~a ~ . ~ '~ '+~ tY1 ~ '~' " i Ll U U W ~ O ~ ~ ~ .-. ~ i .~_O.o~Op~C~ ~~.~- j NE-~p~0.~~a~~~o~
U Wn U ~. C~ ~ ~ y~C~, ~ ~ ~ :.~..' ~ ~ QJ ~O .-, ~ U ~
~~ ~ ~c~ ~'-~O~ > ~ a ~~v~~~y i. ~ '~~'.~ ~,o ~N o o~
v ~.7 L n.a~ ~ ~ ~ o~s'~....
y E.'i y~'~, ~, b1! vl LC ~ ~ ~~ i7.. ~ 'p U N U ~U '~' ~ ~ ~ ~' .'.' W y V ~O
U ~ ~,~, ~ ~ ~y c~' ~ ,~~ ~ ~ O ~ ~ ~ N ~ O W U ~ ~ ~ ~ N O
E-~ O .,.~ ~ at a.., a. U 0.. O ..... ~ tw w .-, U F~ ~ tp C7 ~ ~ ~a ~ U a> L
t~
b v o ~~ o ~°~-''~.~ -d°~"oo '+ ~ bA ~ ~ o . O~ ~ iG O ~ ~ _b,.,U ~ ~
~~r p .~ c~G..O '~ ~ aL7 .fl 4U~
N~ V~1C -0J ~Ut~~"'fli.U.,~,~"'~~~0~ C'.
U ~ C ~r~'. N - ~ .~~' ~ ~ ~y» '~.r U U d N O, ~ '~ _~
0 0 , ~ ~3 ~ ~~~. L ono p. ~ ~ o ~ a. ~ ~ a~
G ~c ~ ~ °' ~ ~~~1 °L' ~ ca ° ~ ~ Y.~ o d cn o .~ ~ ~ ~° : E-, ~c 3 ~-~ b ~
o co M l~ ' op N
v~ O
O
t~ O
p~ N
OO

u., o . ~ ~ o r~
N
~ Q.. a' ~ ~ ' ' ,0~., O
i"' b!1 '-' o I
~ c~ ~ ~ Y "° d P..
v~N~= a~ ~ c.w ~ d~C ' L
~ o o !:
L
7, 7, ~ , U ,.~;

Y ~ ° ~ b Y -~. Y
pCp ° ~ C7 ~ . o ~
N ~ Q ~ ' .,'r-T 4~,Z '*j i~
.p O _ , ~ O ~ O . ' _ ~ ~ ~ ~ _ .
O O
E"'' a b 0 tL0 E-' [-~ ~'u .~ ~"

_ - ~ .b ~ o n. -~' W ~ -d E ~ U ~; ~- ~ c ~ 0 0 ~a ~_ ~ ' ~ o ~ o .

x ~ ~ U
~ U O
~

U '~' U ..57 ~
' ~ cct cC ~ ~
.~ C ~ ~

~~z~ a~~H ~c~a~xr~

U_ ~

'.C7 ~ .~ N 'n b0 > ,"'Y~, ~ ~ ~ N
U ~
' ~

' O N
O - ~ 00 ',~d(1~, 'C3 N ~ O ' ~ CC ~ ~ U *~'' ~ ~ ~ .r U _ ~ U ~
' c ' i a o i o a ~ ~
' ~~
~. o u~ on a~ ~
c r d 'o a~ o ~. U ~

. . ~ ';r .~ ~ O V
~ 'U ~ ~ U ~ ~ ~ ~ ~ O
.

y U V~ 1--;;
v' b0~ ' N
C N ~ U ~ N
~

NLp ' . - O U~"
N ~~~ ~
O~ M
j~~ (%
..~

_ _ ~ ~."' ~O
.
~
~~ CSC

> ~ x .
~ x ~.~
~~ Q ~ o ~.~~:~ a~
~,~ '.aU'~ ~
n odw a .c _~ ~ o o ~~ ;
~ '"' a~ ' U cct t"

~' -' ~. ~ ~, ~'~ ~j fn ..
~ ~i ~ ~~

~_ .'_' ~' ~ ~ .-, ~i-1 ~ ~
~ ..fl ~ ~x~' V7 ~

( ~
> ~~,~.~ ~3~ 3.~~, 3 ~o~M

.~ , .S"., ~~ ~p "L~ r~ ._" ~ fr1 . ~ ~ 'O
~' O

V ~ .O ~ b .C a~ 'O pn N
'o ' 3 ~ ~ x ~~' ~

o ,... .~ ~ $ a.>
,.., ~ ~' d o ~' c o :d ' '~' ~ i ~. ~
O O.. ~ 4..,.-. ~ c7 N U 4: O U
U ~ ~p ~ ~ .~ a". ~
> ~ ~
~ ' t. ..O ~
. ,~ . ~
~ ~ , ,.O ~ bA c ~ ~ ~ ~
~ ~ O O
~ b O A.~
~

." O c v~ O. G +~ O U C
.,~ C O U .~
NN G c ~
~ ~~ D.~ ~' "... ,.
b b NcC A
~ ~~
~

~ .. ?~.
~ ~ ~
~ ~
_ O:.p..,p ~~m ~
~
p ~

U~ - GD~~ 'C3'.C~'~t..~ ~~~
bD,~,~~ ~ b~G~~dJ
~ "f~.

~ ~ ~ ~ ~ C
t" X
p t"
~ '~ ~

.. ~ ~
~' U ~ U
t-, .-.~ U U ~ ,.
.
.~
~ . ~!
~" ~, ~ .~', cn O
~

~O~'~' .~ ~"' U m ~> UO
~ ~O.O~ ~
U pcct0~
~ O U
~
~ ~

. U _ ' +~ C "~ .
.~ v~ *'-, Vi O
bD .~
L T
~O ~
~

' ~~ ~.4:NW n"CI ~~GO~, UO~ O
~~-'j ~

~.~~.~y '~ ~x~d~
.o~U d~
o~~ i~;O~a~;-' . ~,~
~ Ua~4..
"~ ~a~U~
fl~~
~:n' ~~

~az~~~. ,k ,,., a o n"'~
Z . r ~ 3 ~ ' >
~1 ~ >
O
O
' ~' , at O 7 ~..~ N ..., ,_, c , 4. c~ a3 .
..., U ccf U N ..
.. . .
c~ r-..
.
c~

~, O
~ CV

~ d' _ - -, , M
' O

N
U ~ a3 . y .

~ d. . U c~ O ~ ~
N

v~ ~r ~ o ~ en w' ' .
..o U as .
s c~ O
4:
t . ~ . .
~
O ~

O
' ~ N
O
O

O
p ~ 1 ~
c G
cC
Q.. 3 N
j '~ ~ O
V C ~ ~

~ 1 ~ , , C V
bA O ~
N x~. ~
~
~

.
~ ~
N ~, U ~ C := O by :.O
r ~
'~"

U '' l~ CO -te' c ~ ~ ~ ~" ~ ~ , ~
~ ' ~

._ ~ .~ . ,5C J-' Rf .
~, ~ ,.~ ~ U ~ ~
U

p ~ ~ ~~.,, N
~ ~ ~

~ x N ~ . ' p ~ ~ ~ ie .
? o C7 ~ v ~. w' o o ~
~
~ o d r . ~H .
. , .
d ~
M

.~ ~ ~
-~ ~ j ~ ~~o ' > - w ~d ~~~~.'z' o u ~ i a -~

~ .c b > ~" o G~ ~ U ' d o ' ~ ~
d o ~ ~ j~ U
O ~>
~ ~
~

N O M ~C O
~ ~ ~-, ~ N c en a d ~ ~ ~ N ~ ~ M M
~ ~ ~ ~ ~ ~ ~
~

~ ~ . ~ , N > 0 ~, >, U ~

O N ,r o - > ~ .fl ~ Q- O
O ~ ~ .~. ? p ~ O O

O c , , C ~fl d ~'' 'Wy",, -(-'. ~ ~ ~ ~ ;'~ ~", U ~
(.n .. a.r 'y,, - .fl ~ ;
:
~

O 7, O O . .
W~ ~av ~
~ ~ '~ .~.. c. +
->Qdd ~ ~

~xzz , .n o ~ E .x ~.r ~ ~-a ~d ~
o~~-U ~ ~s~va ~o-'-°no~ , .
>. o-o ~ ~
~,a.~~o~Z.~- .
~ .~ ~ ~ ~ ~_ ~ ? .
fn Q, ~ U U ~ H P.
° o -~ ~ ...
3 .~ .o ~ ~ ~ ~,a ~, ~ ~, '"' o ~, 2i o ,", o ~.~ ~ .ia ~.~ a~
.~ ~ > ~x ~ ~ ~o .
.~ ~e o o ~ ~-~
a~ ~ W
y ~
J, C.' ~ ~n N v. .
,~ ~ ,--n . , ~ v ~ ~ ro ~ , cff .~ CC U fr1 ~ a a , ' , 4.i . . ~ . . . . . .
O p U ~
p ~ , ~ ~ o U
p, p .C y cn ~ ~ O > N
V W ~ ~ .~ , O p o m ~ ~ o a) u~ ~ ~ ~b N ~, o 7 ~'~ ~d ~ > .
~, ow -0 ~ Y ~ .
. ~o on r' o .~ a~ o .~_ -c~ ,~ ~-.
,°."~ 'n c~.. "' c ~ ~ p cC > eU ~ ,i~ c~. .~ .~ C;
c~ ~ C .d ~ '~" M p, > ~ c~ .~ N ~ '.~ ~ ;~' O ~ ' " o > °~.3 '~ a. m 'r' ~ o ' ~ ~ ie ~ > ~ o iu n ~°o ~W ~~~~G c~ez ~~'o~j ~ ~w'~ °.o'c ~ ~~ o df~w_ .~ ~s~C.~~E-~~Z_ o~...~U ~ ~ c,> °:U
o ~
.
o ~~ ~~
o ?.+>~..~ ~ o ;w o > ~ ~. _ ~;~ ~ ~OrxU ~ , ~:o a~ ~i-o '~~"~ .
0 0 _ Cj Cj . . ' 59 , .

WO 01/79442 . PCT/USO1/11850 ;_ a~Tnoo~ ~
~~.a'.c~ L d o ~' H ;-o oCadW
E~"'c~°'~.°c~
c~ ~ ~ ..a ~ o ~ 'a...°.~'a~'' °~
~p~dc~px~
o o , -o ~s a~ o >, ° o -o a o ~ o°u~~ .. o.a i U. as ~ ~ (~ b ~ dap ~
cct ° ~ ~ ~ W '-' ~ .C v ~ .~ ~i ~~~.UCn ~, ~ ~ O ~ ° ,-. ° o as >, ° v, y~'~~ wrn~~~~~s.fla3a\.
vQV, ~'o oU ~.c"' a~
~,o t3.~.~y.o ~co ~, ? °' p yn ~ ~ U ~ c c ~ ,n _c~ , ~ ~ .~ V ~d .~' ,.O ~''-~ ~ o at ~
C ~ ' ~ ~1 ~ i ~N ~~" La ~~ 'U-' ~ N N
a y;~ o.~v~~U ~ ~ ~~ ø,~, ~d ~3~N:~ib..DOp..~.fltU.Or~N~~O, O~ ''=' d: O ~' " ~ i1 '~ 01 ..O ~, ' c~ O
.~.LLI~aiN~cC N~....n0>cGAON
n ~-ccf .'. ~ "" 01 ~ O "t".. O O N
U N N ~ 'p b~ L1. ~p U
~ ,..., o ° p o °'.o ~ o ~ >~ >,~HU.n ~
'o ~ ° ~ 0 0 ~ G:d ~ o , oW o ~'n'~ ~,-a ~ ° c.>p ~~~~,x_ _°.o ~ S.' ~ ~ "C3 ~ N a ~ 3 p ~ ~' d. at V V ~ 7."
~~ C1.~ W ~ U H tn y as r... ~ ~. X o ~ .~:. ~ G1 ~ .in.G~1 a~.W, v .~ .b~-~A'ZJ ~' .C ,~
~ Q.N ~~ ~ '~'~ 'C. O, U v '~ ~ b +~
x.~ ~~.~ ~pU ~ ~ ~ ~H.~d.~ ~d ~ ~
CO~O due' N
a1 ~D a' -c''f. 01 O
N
33~
d ~aa~
yd a ~r d ~>N QN
> ~v~ ~a a>~
6; n N ~' ~ N O O ~ O , p d ~ ~
,... a .~ o d .o bll~ ~ ~,.~ ~ N ~ . - .
0v C c~C ~O vyct. C~
N!OO~c.~rOi-T~,~
~~ ~r bD~ U ,~ p U
O ~ °
r~ ~ c~ ~u' ~ ~ d ~ ~ .

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. (See, e.g., the "Biological Activity" and "Therapeutic Protein X" columns 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 reference sequence cited in the "Exemplary Identifier" column of Table 1, and are capable of the therapeutic activity and/or biologic activity of the corresponding, Therapeutic protein disclosed in "Biological Activity"
column of Table 1.
Polypeptide . and Polynucleotide Fragments and Variants Fragments 1S 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.
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, 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 r 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 2S . lacking N-terminal residues of a complete polypeptide retains such immunologi.c activities can readily be determined by routine methods described herein and otherwise known in the art. It is notwnlikely 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.
y 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 (e.g., a Therapeutic protein as disclosed in Table 1). In particular, N-terminal deletions may be described by the general formula m-q, 3S 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), and m is defined as any integer ranging from 2 to q-6. Polynucleotides encoding these polypeptides are also encompassed by the invention.
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). In particular, N
1 ' terminal deletions may be described by the general formula m-585, where 585 is a whole integer representing the total number of amino acid residues in serum albumin (SEQ ID
N0:18), and m iso defined as any integer ranging from 2 to 579.
Polynucleotides encoding these polypeptides are. also encompassed by the invention. ' , ~ 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. In particular, N-terminal deletions may be described by the general formula m-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-6. PolynucIeotides encoding these polypeptides are, also encompassed by the invention.
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 and/or serum albumin protein) 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.
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). In particular, C-terminal deletions may be described by the general formula l-n, where n is any whole integer ranging from 6 to q l, and where q is a whole integer representing the total number of auino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table I).
Polynucleotides encoding these polypeptides are also encompassed by the invention.
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). In particular, C-terminal deletions may be described by the general formula 1-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 serum albumin (SEQ II7 N0:18) minus 1. Polynucleotides encoding these polypeptides are also encompassed by the Invention.
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-n, where n is any whole integer ranging from 6 to q-l, 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.
In addition, any of the above described N- or C--terminal deletions can be combined to produce a N- and C-terminal deleted reference polypeptide. The invention also provides 1 S 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-n of a reference polypeptide (e.g., a Therapeutic protein referred to in Table l, or senim albumin (e.g., SEQ
ID N0:18), or an albumin fusion protein of the invention) where n and m are integers as described above. PolynucIeotides encoding these polypeptides are also encompassed by the invention.
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, serum albumin protein or ari albumin fusion protein 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.
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. , 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 piesent invention. The biological activity of the fragments may include, an improved desired ,activity, or a decreased undesirable activity.

WO 01/79442 , PCT/USO1/11850 Variants "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.
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 differing in sequence from a Therapeutic protein (e.g.
10. see "therapeutic" column of Table I), albumin protein, and/or albumin fusion protein of the invention, respectively, but retaining at least one functional and/or therapeutic property thereof (e.g., a therapeutic activity and/or biological activity as disclosed in the "Biological Activity" column of Table 1) as described elsewhere herein or otherwise known in the art.
Generally, variants are overall very similar, and, in many regioils, identical to the amino acid sequence of the Therapeutic protein corresponding to a,Therapeutic protein portion of . an albumin fusion protein of the invention, albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention, and/or albumin fusion protein of the invention. Nucleic acids encoding these variants are also encompassed by the invention.
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., an amino acid sequence disclosed in the "Exemplary Identifier" coluzrin of Table l, or fragments. or variants thereof), albuW in proteins (e.g., SEQ ID
N0:18 _or fragments or variants thereof) corresponding to an albumin protein portion of an albumin fusion protein of the invention, and/or albumin fusion proteins of the invention. 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 amino acid sequence 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 O.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, AusubeI, 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 arer also encompassed by the invention. -By a polypeptide . having an amino acid sequence at least, for example, 95%
"identical" to a query amino acid sequence of the present invention, 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 S% 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.
As a practical matter, whether any particular polypeptide is at least 80%, 85%o, 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 the Therapeutic protein portion of the albumin fusion protein or the .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=l, 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.
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-terminal truncations of the subject sequence when calculating global percent identity. For subject sequences truncated at the N- and C-termini, 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.
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 matchedltotal 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.
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-Ievel 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-X00 (1993), fully incorporated by reference) which are tailored for sequence similarity searching. ' ' . .
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 WO 01/79442 . PCT/USO1/11850 similarity searching of sequence databases, see Altschul et al., (Nature Genetics 6: 119-129 (1994)) which is fully .incorporated by reference. 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), fully incorporated by reference). 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 foi 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 wink'h 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=l; 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.
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 f-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 colon expression for a particular host (change ~codons in the human mRNA to those preferred by a bacterial host, such as, yeast or E. coli).
In a preferred embodiment, a polynucleotide encoding an albumin portion of an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells. In further preferred. embodiment, a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein, of the invention is optimized for expression in yeast or mammalian cells. In a still further preferred embodiment, a polyni~cleotide encoding ~an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
In an alternative embodiment, a colon optimized polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein of the invention does not hybridize to the wild type poIynucleotide ,encoding the Therapeutic protein under .
stringent hybridization conditions as described herein. In a further embodiment, a colon optimized.

polynucleotide encoding an albumin portion of an albumin fusion protein of the invention 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 encoding an albumin fusion protein of the invention does not hybridize to the wild type polyriucleotide encoding the Therapeutic protein portin or the albumin protein portion under stringent hybridization conditions as described herein.
In an additional embodiment, polynucleotides encoding, a Therapeutic protein portion T
of an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of that Therapeutic protein. In a further embodiment, polynucleotides.encoding an albumin protein portion of an albumin fusion protein of the invention, do not comprise, or alternatively consist of, the naturally occurring sequence of albumin protein. In an alternative embodiment, polynucleotides encoding an albumin fusion protein- of the invention do not comprise, or alternatively consist of, of the naturally occurring sequence of a Therapeutic protein portion or the albumin protein portion.
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:Il, 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. ' 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. 268:12984-2988 (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).) Motreover, 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 signif cantly differed in activity from wild-type. , 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.
Thus, the invention further includes polypeptide variants which have a functional activity (e.g., biological activity and/or therapeutic activity).. In highly preferred embodiments the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity, such as that disclosed ,15 . in the "Biological Activity" column in Table 1) 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.
. 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 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.
. -, 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 df an amino acid sequence to change.
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.
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. , 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 Fe fusion region peptide, . - Such variant polypeptides are deemed to be .within the scope of those skilled in the art from the teachings herein.
For example, polypeptide. variants containing amino acid substitutions of charged amino acids with other charged or neutral. amino acids may produce proteins with improved 30. 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-( 1993).
In specific embodiments, the polypeptides of the invention comprise, or alternatively, consist.of, fragments or variants of the amino acid sequence of a. Therapeutic protein described herein and/or human serum albumin, and/or albumin fusion protein of the WO 01/79442 ~ PCT/USO1/11850 invention, wherein the fragments or variants have 1-5, 5-10, S-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.
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 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 (I992)).
Functional activity "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 Iigand for a polypeptide. , "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 2S-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).
In preferred embodiments, an albumin fusion protein of .the ~ invention has at least , one biological andlor therapeutic activity associated with the Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin. .
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. Specifically, albumin fusion proteins may be assayed for functional activity (e.g., biological activity or, therapeutic activity)' using the assay referenced in the - "Exemplary Activity Assay" column of Table 1. Additionally, one of skill in the art may routinely assay fragments of a Therapeutic protein corresponding to a Therapeutic protein ' portion of an ~aIbumin fusiom protein of the invention, for activity using assays referenced in its corresponding row 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 awalbumin b fusion protein of the invention, for activity using assays known in the art' and/or as described in the Examples section below.
. For example, in one embodiment where one is assaying for the ability of an albumin fusion protein of the invention 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, enzynrie 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.
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 containing 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-I23 (1995). In another embodiment, the ability of physiological correlates of an albumin fusion protein of the present invention to bind to a substrates) of the Therapeutic polypeptide corresponding to the Therapeutic portion of the albumin fusion .protein of the invention can be routinely assayed using techniques known in the art. .
~In an alternative embodiment, where the ability of an albumin fusion protein of the invention to multimerize is being evaluated; association with other components of the ~20 ~ 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.
In preferred embodiments, an albumin fusion protein of the invention comprising all or a, portion of an antibody that bids 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 of the invention 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 polypeptide that is specifically bound by antibody that binds a .
Therapeutic protein.
Albumin fusion proteins of the invention (e.g., 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 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 Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques 'known in the art.
Assays for the ability of the albumin fusion proteins of the invention (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 (199I)), 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)) (each of these references is. incorporated herein in its entirety by reference). Albumin fusion proteins of the invention 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.
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 - molecules) 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.
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. l;
John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety). Exemplary immunoassays are described briefly below (but are not intended by way of limitation): . .
, Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer ( 1 % NP-40 or Triton X-100, 1 % sodium deoxycholate, 0.1% SDS, 0.15 M -NaCI, 0:01 M ,sodium phosphate at pH 7.2, 1% Trasylol) WO 01/79442 , PCT/USO1/11850 supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, 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 tirrie (e.g., 1 to 4 hours) at 40 degrees C, adding sepharose 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 of the invention 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 sepharose beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol..l, John Wiley & Sons, Inc., New York at 10.16.1.
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-Tween 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., 3zP or lzsl) 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 ),0.8.1.
ELTSAs 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 ~~
. 75 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. l, John Wiley & Sons, Inc., New York at 11.2.1.
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 'zsI) 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 of the present 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 albumin fusion protein, can also be determined using radioimmunoassays. In this case, the protein, antigen or epitope is incubated with an albumin fusion protein of the present invention conjugated to a labeled compound (e.g., 3H or'zsI) 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.
In a preferred embodiment, BIAcore kinetic analysis is used to determine the binding on and off hates 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.
Antibodies that bind a Therapeutic protein corresponding to the Therapeutic protein portion of an albumin fusion protein of the invention 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-z M, 10-z M, 5 X 10-3 M, 10-3 M, 5 X 10-4 M, 104 M.
More preferred binding affinities include- those with a dissociation constant or Kd less than 5 X

105 M, 10-5 M, 5 X 10-6 M, 10-6M, 5 X 10-' M, 10' M, 5 X 10-$ M or 10~$ M.
Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X
109 M, 10-9 M, 5 X 10-'° M, 10-'° M, 5 X 10-" M, 10-'1 M, 5 X 10-12 M, 'o-iz M, 5 X 10n3 M, 10''3 M, 5 X 1014 M, 10-'4 M, 5 X 10~" 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 of the present invention 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 of the present invention. Other methods will be known to the skilled artisan and are within the scope of the invention.
Albumin 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 or chemical conjugation. : .
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).
As used herein, "albumin" refers collectively to albumin protein or amino acid sequence, or an albumin fragment or variant, having one or more functional activities (e.g., biological activities) of albumin. In particular, "albumin" refers to human albumin,or fragments thereof (see EP 201 239, EP 322 094 WO 97/24445, W095/23857) especially the mature form of human albumin as shown in Figure 15 and SEQ ID N0:18, or albumin from other vertebrates or fragments thereof, or analogs or variants'of these molecules or fragments thereof. ~ ' 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 ID N0:18: 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 Gln, and Lys-414' to Gln (see, e.g., International Publication No. W095/23857, hereby incorporated in its entirety by reference .
herein). 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.
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 as shown in Figure 15, 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.
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 rion 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.
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. , ' 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 N0:18), .2 (amino acids 195-387 of SEQ ID N0:18), 3 (amino acids 388-585 of SEQ ID N0:18), 1 + 2 (1-387 of SEQ ID N0:18), 2 + 3 (195-58S of SEQ ID N0:18) or 1 + 3 (amino acids 1-194 of SEQ ID N0:18 + amino acids 388-585 of SEQ ID N0:18).
Each domain is itself made up of two homologous subdomains namely 1-105, I20-194, 1-95-291, 316-387, 388-491 and 5I2-585, with flexible inter-subdomain linker regions comprising residues Lys106 to G1u119, G1u292 to Va1315 and G1u492 to A1a511.
~ 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 Specifically bind Therapeutic proteins are also Therapeutic proteins 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 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 an antibody that binds a Therapeutic protein.
Antibody structure and background The basic antibody structural unit is known to comprise a tetramer. Each tetramer 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 of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines Ja constant.region primarily responsi-ble'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, lgG, IgA,.and IgE, respectively.
See generally, Fundamehtal Immunology Chapters 3-5 (Paul, W., ed., 4th ed.
Raven r 'Press,.N.Y. (1998)) (incorporated by reference in its entirety for all purposes). The variable regions of each lightlheavy chain pair form the antibody binding. site.
Thus, an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same.
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 arid 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 FR1, 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 definitions 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).
As used herein, "antibody" refers to iW munoglobulin 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 chirrieric 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 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. ' .
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. ' .
. 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, lgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, Ig"G2, 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 of the invention are IgGl. 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 of the invention 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 of the invention are IgG4.
Most preferably 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 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 re~gion(s) alone or in~ combination with the entirety or a portion of the following: hinge region, CHl, CH2, and CH3 domains.
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 monospecific, bispecific, trispecific or of greater multispecificity.
Multispecific antibodies may be specific for different epi~topes 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 91100360; WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S.
Patent Nos. 4,47.4,893; 4,714,681; 4,925,64f; 5,573,920; 5,601,819; Kostelny et al., J.
Immunol. 148:1547-1553 (1992). ' Antibodies that bind a Therapeutic protein (or fragrrient 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. "'Diabodies': 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" Irct J Cancer Suppl 7:51-52 (1992)).
The present invention also provides albumin fusion proteins that comprise, fragments or variants (including derivatives) of an antibody described herein ox 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 subsitutions, 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 VH
domain, VHCDR1, VHCDR2, VHCDR3, VL domain, VLCDRl, 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.
Antibodies thatbind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion 'protein of the invention may be described or specified in terms of the epitope(s) or portions) 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 corresponding antibody (not fused to albumin), that binds a Therapeutic protein.
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 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%
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 of the invention 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%
identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also , included in the present invention. Iri a specific embodiment, the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic WO 01/79442 - . PCT/USO1/11850 polypeptide, or combinations) 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 corresponding antibody (not fused. to albumin) that binds a Therapeutic protein.
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-z M, S X 10-3 M, IO'3 M, 5 X
10-4 M, 10-4 M. More preferred binding affinities include those with a dissociation constant or Kd less than S X 10-5 M, 10-S M, 5 X 10-6 M, 10-6M, S X IO'' M, 10' M, 5 X
10-fi M or 10-8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than S X 10-9 M, 10-9 M, 5 X 10-'° M, 10-1° M, 5 X 10-1' M, 10-11 M, 5 X 10''2 M, l0-12 M, 5 X 10-13 M, 10-13 M, 5 X 10-'4 M, 10-1'' M, 5 X 10-'S 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.
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 8S %, 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 an antibody to an epitope of a Therapeutic protein as well as the corresponding antibody (not fused' to albumin) that binds a Therapeutic protein, competitively inhibits binding of an 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 the corresponding antibody (not fused to albumin) that binds ~a Therapeutic protein 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%.
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 immurioprecipitation 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 the corresponding antibody (not fused to albumin) that binds a Therapeutic protein.
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 ligarld-mediated receptor activation, for example, by inducing dimerization of the receptor. The antibodies may be specif ed as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the Therapeutic protreins (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. Immunol.
161(4):1786-1794 (1998); Zhu et al., Cancer Res: 58(15):3209-3214 (1998); Yoon et al., J. Immunol. 160(7):3170-3179 (1998); Prat et al., J. Cell. Sci. 111(Pt2):237-247 (1998);

Pitard et al., J. Immunol. Methods 205(2):177-190 (1997); Liautard et al., Cytokine 9(4):233-241 (1997); Carlson et al., J. Biol. Chem. 272(17):11295-1,1301 (1997); Taryman et al., Neuron 14(4):755-762 (1995); Muller et al., Structure 6(9):1153-1167 (I998);
Bartunek et al., Cytokine 8(1):14-20 (1996) (which are all incorporated by reference herein ~ in their entireties). 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 the corresponding antibody that binds a Therapeutic protein not fused to albumin.
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 ih 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); incorporated by reference herein in its entirety. 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 in vitro and i~ vivo diagnostic and therapeutic methods.
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, phosphylation, 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, belt not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tuiticamycin, 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 Producihg Antibodies that bind Therapeutic Proteins 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.
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.
I988); Hammerling, et al.; in: Monoclonal Antibodies and T-Cell Hybridomas 563-, (Elsevier, ~N.Y., 1981) (said references incorporated by reference in their entireties). 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.
Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. In a nori-limiting example, mice can be immunized with a Therapeutic protein or fragment or variant thereof or a cell 'expressing such a Therapeutic protein or fragment or variant thereof. 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. ~ Hybridomas 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.
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 3S splenocytes isolated from, a mouse immunized with ari 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.

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, which is hereby incorporated in its entirety by reference.
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.
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 t 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.
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 immyoglobulin 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 xegion and the CHl domain of the heavy chain.
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 III 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 al., 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 95115982; 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; each of which is incorporated herein by reference in its entirety.
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(ab')2 fragments can,also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques 12(6):864-869 (1992); and Sawai et al., AJRI 34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988) (said references incorporated by reference in their entireties).
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 al., 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 immunoglobulin 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., (~I989) J. Immunol. Methods 125:191-202; U.S. Patent Nos.

5,807,715; 4,816,567; and 4,816397, which are incorporated hereim by reference in their entirety. 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 arid 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; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Antibodies can be -hurizanized 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;
Padlan, 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).
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, W0 98/50433, WO 98/24893, WO 98/16654-, WO
96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety. , Human antibodies can also be produced using transgenic mice which are incapable of expressing functional, endogenous immunoglobulins, bLlt which can express human immunoglobuliri 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 immurioglobulin 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 nuce.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, which are incorporated by reference herein .in their entirety. 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. - _ 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., Biotechnology 12:899-903 (1988)).
PolyaLCeleotides Encoding Antibodies 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 sacpra, to polynucleotides that encode an antibody, preferably, that specifically binds to a Therapeutic protein, preferably, an antibody that ~ binds to a polypeptide having the amino acid sequence of a "Therapeutic Protein X" as discosed in the "Exemplary Identifier" column of Table 1.
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 WO 01/79442 , PCT/USO1/11850 ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR. ' _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 eDNA 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 60).
Once the nucleotide sequence and . corresponding airiino 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 8r. Sons, NY, which are both incorporated by reference herein in their entireties ), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, ~ and/or insertions.
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 WO 01/79442 . PCT/USO1/11850 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.
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 immunoglobulin constant region, e.g., humanized antibodies.
Alternatively, techniques described for the production 'of single chain antibodies (U.S. Patent No. 4,946,778; Bird, Science 242:423- 42 (I988); 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 et al., Science 242:1038- 1041 (1988)).
Recombinant Expression of Antibodies 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 polynucleodde 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 franscriptional and translational control signals. These methods include, fox example, in vitro recombinant DNA,techniques, synthetic techniques, and in wivo 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 CT.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.
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.
A 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 calls, 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)).
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 pUR278 (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,; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chem. 24:5503-5509 (1.989)); 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 adsofption 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.
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).
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 Bitiner et al., Methods in Enzymol. 153:51-544 (1987)). ' 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 bf the protein. Different host cells have characteristic and specific mechanisms for the post-transIational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to , ensure the correct modification and processing bf 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, BHK, 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.
For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell Iines~ 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 engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
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 phosphoribosyltrailsferase (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., Pioc.
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 aminoglycoside G-418 Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Pharmacol.~ Toxicol. 32:573-(1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann.
Rev.
Biochem. 62:191-217 (.1993); May, 1993, TIB TECH 11(5):155-21-5 (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 al. (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), which are incorporated by reference herein in their entireties.
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 amplif cation 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 (Grouse et ~al., Mol. Cell. Biol. 3:257 (1983)).
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 availabilty of cell lines (e.g., the murine myeloma cell line, NSO) i-vhich are glutamine synthase negative.
Glutamiize 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;
~ o W089/01036; W089/10404; and W091/06657 which are incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors that may be used according to the present invention are'commercially available from suppliers, including, fox 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., Bioltechhology 10:169(1992) and in Biblia and Robinson Baotechnol.
Prog. 11:1 (1995) which are incorporated in their entirities by reference herein. .
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 light chains may comprise cDNA or genomic DNA.
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. .
Modifieations of Antibodies 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 "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767. (1984)) and the "flag" tag.
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 nonradioa~tive 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 WO 01/79442 ~ PCT/USO1/11850 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 125I, 131I, Z llIn or 99Tc. Other examples of detectable substances have been described elsewwhere 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, antimetabolifes (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).
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, l3-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 9?/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et cal., Int. Immurcol., 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 ("IL-1"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ~~"GM-CSF"), granulocyte y colony stimulating factor ("G-CSF"), or other growth factors.
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.
Techniques for conjugating such therapeutic moiety to antibodies are well known.
See, for example, Arnon 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 . 15 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", Immunol. Rev. 62:119-58 (1982).
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, which is incorporated herein by reference in its entirety.
An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factors) and/or cytokine(s) can be used as a therapeutic.
Antibody-albumin ' fusion ' .
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.
In specific embodiments, the fragment or variant of an antibody that ~
specifically ' binds a Therapeutic protein and that corresponds to a Therapeutic protein portion , of an~
albumin fusion protein comprises, or alternatively consists of, the VH domain.
In other embodiments, the fragment or variant of an antibody that specifically 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 specifically 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 specifically 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 specifically 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.
.. In specific embodiments, the fragment or variant of an antibody that specifically 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 specifically 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 specifically 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 specifically 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 specifically 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.
In other embodiments, the fragment or variant of an antibody that specifically 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.
In preferred embodiments, the fragment or variant of an~antiliody that specifically 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 N0:36).
Immunophenotyping 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 . . 100 antibodies (or albumin fusion proteins comprsing 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)).
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 Uariant~of an Antibody that binds a Therapeutic ' Protein 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. .
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)) (each of these references is incorporated herein in its entirety by reference). 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.
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 molecules) 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.
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, radioimmurioassays, ,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, which is incorporated -by reference herein in its entirety)'. Exemplary immunoassays are described briefly below (but are not intended by . way of limitation).
Immunoprecipitation protocols generally comprise lysing, a population of cells in a~
lysis buffer such as RIPA~buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1 % SDS, b:15 M NaCI, 0.01 M sodium phosphate at.. pH -7.2, 1 % Trasylol) supplemented with protein phosphatase andlor 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 sepharose beads (or beads coated with an appropriate anti-iditoypic 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 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 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. l, John Wiley & Sons, Inc., New York at 10.16.1.
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-Tween 20), 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 antibody or 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'~'I) 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 arid 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 weld 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. l, John Wiley & Sons, Inc., New York at 11.2.1.
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 '25I) 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 present 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 present 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 epuitope as the albumin fusion protein of the invention. ' 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. BIAcore 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.
Therapea~tic Uses The present invention is further directed to antibody-based therapies which involve administering antibodies of the inventibri 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 --'~"°"'~ al least a fragment or variant of an antibod tha WO 01/79442 be used to treat, inh' .
AAi°idpeutic protein can ibit or prevent d Y t binds associated with aberrant expression and/or activi of lseases, disorders or conditions not limited to, any one or more of the diseases, disor tY a Therapeutic protein, including, but S The treatment and/or prevention of disease tiers, ox conditions described herein.
aberrant expression and/or activity of a Thera cut' s, disorders, or conditions associated wit h alleviating symptoms associated with those p is protein includes, but is not 1i mited to, the invention or albumin fusion proteins of the in diseases, disorders or conditions. antibodies of variant o~an antibody that binds a Therapeutic r vention comprising at least a fra en t~ t or acceptable compositions as known in the art p otein may be provided in pharmac eutically or as described herein.
In a specific and prefer-ed embodiment, the rese based therapies which involve administering antib p tit invention is directed to antibod Y
proteins of the invention comprising at least a fragment odies of the invention or albumin fusion Therapeutic protein to an animal, preferabl a or variant of an antibody that b' lads a 1 S . patient for treating one or more diseases, dis Y mammal, and most preferabl a Y human, to: neural disorders, immune system disorders, musc orders, or conditians, including but not limited gastrointestinal disorders, pulmonary disorders, cardio ular disorders, reproductive disorders,.
proliferative disorders, andlor cancerous diseases a vascular disorders, renal disorders nd conditions ' elsewhere herein. Therapeutic compounds of the i and/or as described ~0 antibodies of the invention (e.g., an~bodies nvention include, but ~e not 1i mited to, the cell surface °f a mammalian cell; antibodies directed to the full length protein expressed o n protein and nucleic acids encoding antibodies of the inv directed to an epitope of a Therapeutic and derivatives thereof and anti-idiotypic antibodies a ention (including fragments, analo s g the invention can be used to treat, inhibit s described herein). The antibo ' dies of ZS associated with aberrant expression and/or a or prevent diseases, disorders or co nditions not limited toy any one ox more' of the diseases, disor ct'vI~' °f a Therapeutic protein, including, but The treatment andlor prevention , of di tiers, or conditions descri bed herein.
seases, disorders, or conditions associated with a errant expression andlor activity of a Therapeutic ro alleviating symptoms associated with those disease p tein includes, but is .not limited to 30 the invention. or albumin fusion proteins of s, disorders or conditions. Antib odies of variant of an antibody that. binds a Therapeutic rotei the invention comprising at least a fragment ox acceptable compositions as known in the art or as descri p n may be provided in pharmaceuti call Y
bed herein.
A summary of ~e ways in which the antibodies ~ . ' proteins of the invention comprising at least a fragment of the, invention or albumin fusion 3S Therapeutic protein may be used thera a or variant of an antibody that binds a locallyor systemically in the body or by direct c t p utically includes binding Therapeutic rotei P ns bY complement (CDC) or by effector cells y otoxicity of the antibody, e.g. as mediate d . (ADCC)~ Some of these approaches ~ are ..
lOS -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.
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.
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, horrrional 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.
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 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 X 10-6 M, 10-6M, 5 X 10-' M, 10' M, 5~ X 10-g M or 10-$ 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 I0-'°'M, 10-'° M, 5 X 10-'2 M, 10-" M, 5 X 10-12 M, '0-'2 M, 5 X 10-'3 M, 10-'3 M, 5 X 10-'" M, 10-''' M, 5 X 10-'S M, or 10-'s M. .
Gene Therapy . ' In a specific embodiment, nucleic acids comprising sequences encoding antibodies that bind Therapeutic proteins or albumin fusion proteins comprising at least a fragment or varaint 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.
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 The compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in viva 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 cultuxe, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed.
TherapeuticlProphylactic Administration and Composition 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, preferably an antibody. 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.
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 arid routes of administration can be selected from among those described herein below.
, 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 WO 01/79442 ~ PCT/USO1/11850 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 intradermal, 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 Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
In a specific embodiment, it may be desirable to administer ahe 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. ' In another embodiment, the compound or composition can be delivered in a vesicle, in particular a Iiposome (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, Nevv York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.) 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.Neurosnrg. 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. 11S-138 (1984)).
Other controlled release systems are. discussed in the review by Langer (Science 249:1527-1533 (1990)).
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 .enfer 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 1S incorporated within host cell DNA for expression, by homologous recombination.
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 "earner" 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 2S 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 3S 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 _ . r 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.
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 Iocal anesthetic such as Iignocaine 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.
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.
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.
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 arid tissue penetration.
(e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.
Diagnosis aid Imaging Labeled antibodies and derivatives and analogs thereof that bind a Therapeutic protein (or fragment or variant thereof) (including albumin fusiori 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. , 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.
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 irrimunohistological 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 (1-987)). 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 (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
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.
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 fragement 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 andvB. A. Rhodes, eds., Masson Publishing Inc. (1982)).
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.
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:

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), arid sonography.
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).
Ki is 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, iri 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).
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 may include 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 WO 01/79442 . . PCT/USO1/11850 chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support.
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.
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 b~e 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..
In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by fhe 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 xeagent 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).
. 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).
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 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 polyriucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or chemical conjugation to one another. The Therapeutic protein and albumin protein once part of the albumin fusion protein, may be referred to as a. "portion", "region" or "moiety" of the albumin fusion protein.
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. Iri 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.
In further embodiments, the invention provides an albunnin fusion protein comprising, or alternatively consisting of, a Therapeutic protein,, and a biologically active andlor 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 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.
Preferably, the albumin fusion proteiwcomprises 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.
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 a . 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 disease,~disorder, or condition (e.g. as listed in the "Preferred Indication Y" column of Tabled). 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 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.
In addition to albumin ~ fusion protein in which the albumin portion is fused N
terminal andlor 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 diclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof) into an internal region of I=IA. 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 (see Figures 9-11).
The loops, as determined from the crystal structure of HA (Fig. 13) (PDB
identifiers 1A06, IBJS, IBI~E, 1BM0, IE7E to lE7I and IUOR) 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.
Loops in human albumin structure into which peptides or polypeptides may be inserted to generate albumin fusion proteins of the invention include: Va154-Asn6l, Thr76-Asp89, A1a92-G1u100, G1n170-A1a176, His247-G1u252, GIu266-GIu277, G1u280-His288, A1a362-GIu368, Lys439-Pro447,Va1462-Lys475, Thr478-Pro486, and Lys560-Thr566. In more preferred -embodiments, peptides or polypeptides are inserted into the Va154-Asn6l, G1n170-A1a176, and/or Lys560-Thr566 loops of mature human albumin (SEQ ID N0:18).
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.

WO 01/79442 . PCT/USO1/11850 Such library(s) could be generated on HA or domain fragments of HA by one of the -following methods:
(a) 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 (for example see Fig. 10a);
(b) replacement of,. or insertion into one or more loops of HA or HA domain fragments (i.e., internal fusion) of a randomized peptides) of length Xn (where X is. an amino acid and n is the number of residues (for example see Fig. 10b);
(c) N-, C- or N- and C- terminal peptide/protein fusions in addition to (a) and/or (b)..
The HA or HA domain fragment may also be made multifunctional by grafting the peptides derived from different screens of different loops against different targets into the same HA or HA domain fragment. .
In preferred embodiments, peptides inserted into a loop of human serum albumin are peptide fragments or peptide variarits of the Therapeutic proteins disclosed in Table 1. More particulary, 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 1 l, 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 iri 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 1 l, at least I2, at least 13, at least I4, 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 25- 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.
Generally, the albunnin 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.
For example, an anti-BLyST"' 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-IFNa.-2b fusion mixed with HA-anti-BLyST"' scFv fusion or other HA-fusions in various ratio's depending on function, half life etc.
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.
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 Xn (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.
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.
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.
Therefore, as described above, the albumin fusion proteins of the invention may have the following formula R1-L-R2; R2-L-Rl; or R1-L-R2-L-Rl, wherein R1 is at least _ 1 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. .
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.
. ~ 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, S 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.
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 l, an 1S 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 S weeks or more under various temperature conditions.
Expression of Fusion Proteins 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.
We have found that, by fusing the hGH coding sequence to the HA coding sequence, either to the 5' end or 3' end, it is possible to secrete the albumin fusion protein from yeast without the requirement for a yeast-derived pro sequence. This was surprising, as other workers have found that a yeast derived pro sequence was needed for efficient secretion of hGH in yeast.
For example, Hiramatsu et cal. (Appl Environ Microbiol 56:2125 (1990); Appl Environ Microbiol 57:2052 (1991)) found that the N-terminal portion of the pro sequence in the MLacor pzssillacs rennin pre-pro leader. was important. Other authors, using the MFa-1 signal, have always included the~MFa-1 pro sequence when secreting hGH. The pro sequences were believed to assist in the folding of the hGH by acting as an intxamolecular chaperone. The present invention shows that HA or fragments of HA can perform a similar ~furiction. c Hence, 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 WO 01/79442 ~ PCT/USO1/11850 the fused molecule of the first aspect of the invention, there being no yeast-derived pro sequence between the signal and the mature polypeptide.
The Saecharornyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence. - ~ ' ~ 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.
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, I~luyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
Preferred yeast strains to be used in the production of albumin fusion proteins are .
D88, DXY 1 and BXPIO. D88 [leu2-3, leaa2-122, canl , pral , ubc4j, is a derivative of parenf strain AH22his+ (also known. as ,DB1; see, e.g., Sleep et al. 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 PRB 1 in glucose excess. The PRB 1 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 D88 exhibits the repression by glucose but maintains the induction upon entry into stationary phase. The PRAT 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 shbrt~
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. W0991b0504, hereby incorporated in its entirety by reference herein).
DXY,1, a derivative of D88, has the following genotype: [leLa2-3, leu2-122, canl , pr-al, ubc4, acra3::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, KK) 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 Derry-Williams et al., Yeast 14:161-169 (1998), . hereby incorporated in their entireties by reference herein).
BXP10 has the following genotype: leu2-3, leu2-122, canl, pral, ubc4, ura3, yap3:: URA3, lys2, hspl SO::LYS2, pmtl:: URA3. In addition to the mutations isolated in DXY1, this strain also has a knockout of the PMT1 gene and the HSP150 gene.
The PMTI gene is a member of the evolutionarily conserved family of dolichyl-phosphate-D
.~ mannose protein O-mannosyltransferases (Pmts). The transmembrane topology of Pmtlp suggests that it is an integral membrane protein of the endoplasmic reticulum with a role in O-linked glycosylation. This mutation serves to reduce/eliminate O-linked glycosylation of HSA fusions (see, e.g., International Publication No. W000144772, hereby incorporated in its entirety by reference herein. 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, hereby incorporated in its entirety by reference herein.
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. . .
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. (I985) Biotech. 3, 208. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
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, L,EU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
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 2. Figure 4 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 (PRBlp), a Fusion leader sequence (FL), DNA
encoding HA (rHA) and an ADHI 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 albumiw (SEQ ID N0:29) and the last five amino acids of the mating factor alpha 1 promoter (SLDKR, see EP-A-387 319 which is hereby incorporated by reference in its ehtirety.
_The plasmids, pPPC0005, pScCHSA, pScNHSA, and pC4:HSA were deposited on April.ll, 2001 at the American Type Culture Collection,. 10801 University Boulevard, Manassas, Virginia 20110-2209 and given accession numbers ATCC , , , and , respectively: Another vector useful for expressing an albumin fusion protein in yeast the pSAC35 vector which is described in Sleep et al., BioTechnology 8:42 (1990) which is hereby incorporated by reference in its entirety. .
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.
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, = single-stranded termini with their 3' 5'-exonucleolytic activities, and fill in recessed 3'-ends with their polymerizing activities.
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.
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. ' 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 . Saiki et al. ( 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.
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 (formerly classified as Hansenula), Saccharomyces, Kluyveromyces, Aspergillus, Cafidida, Torulopsis, Torulaspora, Schizosaccharomyces, . Citeromyces, Pachysolen, Zygosaccharomyces, Debaromyces, Trichodernaa, Cephalosporium, Humicola, Mucor, Neurospora, Yarrowia, Metschunikawia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Piclzia and Torulaspora.
Examples of Saccharomyces spp. are S. cerevisiae; S. italicus and S. rouxii.
Examples of Kluyveromyces spp. are ,K. fragilis, K. lactic and K. marxianus. A
suitable Toraclaspora species is T. delbrueckii. Examples of Pichia (Hansenula) spp. are P.
aregusta (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, all of which are incorporated herein by reference.
Preferred exemplary species of Saccharomyces include S. cerevisiae, S.
italicus, S .
diastaticus, and Zygosaccharomyces rouxii. Preferred exemplary species of Klaayveromyces include K. fragilis and K. lactic. 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.
pastoris.
. Preferred exemplary species of Aspergillus include A. niger and A. nidulans.
Preferred exemplary species of Yarrowia 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); Saccharornyces cer-evisiae 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, teIeomorph (ATCC Accession No.
62987); Kluyveromyces lactic (Dombrowski) van der Walt, teleomorph (ATCC, Accession No. 76492); ~Pichia angusta (T'eunisson et al.) Kurtzman, teleomorph deposited as Hansenula polymorpha de Morais et Maia, teleomorph (ATCC Accession No. 26012);
Asper~gillus rciger van Tieghem, anamorph (ATCC Accession No. 9029);
Aspergillus niger van Tieghem, .anamorph (ATCC Accession No. 16404); Aspergillus nidularcs (Eidam) Winter, anamorph (ATCC Accession No. 48756); and Yarrowia lipolytica (Wickerham et .al.) van der Walt et von Arx, teleomorph (ATCC Accession No. 201847).
Suitable promoters for S. cerevisiae include those associated with the PGKI
gene, GAL1 or GAL10 genes, CYCI, PHOS, TRPI, ADHI, ADH2, the genes fox glyceraldehyde-3-phosphate dehydrogenase, hexokinase, 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).
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 ~iepressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
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, incorporated herein by reference), and Pichia expression kits are commercially available from Invitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, California. Suitable promoters include AOXI and AOX2. 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 KI
uyueromyces spp., a ' suitable promoter being PGI~I.
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.
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 S .
cerevisiae include that from the mating factor alpha polypeptide (MFa.-1) and the hybrid leaders of EP-A-387 319. Such leaders (or signals); are cleaved by the yeast before the mature albumin is released into the~surrounding medium. Further. such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 911036516), acid phosphatase (PH05), the pre-sequence of MFa-I, 0 glucanase (BGL2) and~killer toxin; S.
124 ' daastaticus glucoarnylase Il; S. carlsbergeresis a-galactosidase (MELD; K.
lactic killer toxin;
and Candida glucoarnylase.
Additional Methods of Recombinant and Synthetic Production of Albumin Fusion Proteins 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.
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.
The polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lae, trp, p7aoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, 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.
As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G41S, glutamine syrithase, 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.
Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors; pNHBA, pNHI6a, pNH 18A, pNH46A, available from Stratagene Cloning Systems, , Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRITS available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTI and' pSG
available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
Preferred expression vectors for use in yeast systems include, but are not limited to .pYES2, pYDI, pTEFI/Zeo, pYES2lGS; pPICZ, pGAPZ, pGAPZaIph, 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.
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. coli, ,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 localizatiori 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, the contents -of which are herein incorporated by reference in their entireties.
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, the MPIF-I signal sequence (e.g., amino acids 1-2I of GenBank Accession number AAB51134), the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, ~ SEQ ID
N0:34),~ and a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID
N0:35). A suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1-19 of GenBank Accession Number AAA72759).
~ 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 availabilty of cell lines (e.g., the marine 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: W087/044.b2;
W086/05807;
W089/01036; W089/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. 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 marine myeloma cells is described in Bebbington et al., Bioltechnology 10:169(1992) and in Biblia and Robinson Biotechreol.
Prog. 11:1 (1995) which are herein incorporated by reference.
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.
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 aie described in' many standard laboratory manuals, such as Davis etaL, Basic 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. ~ ' 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.
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. Sci. USA 86:8932-8935 (1989);
and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
Albumin fusion proteins , of the invention can be recovered and purified from I5 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. .
In preferred embodiments the albumin fusion proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAF . sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
In specific embodiments the albumin fusion proteins of the invention are purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM
sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S
and CM, Fractogel S and CM columns and their equivalents and comparables. v ' 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-sepharose, '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.
In specific embodiments the albumin fusion proteins of the .invention are purified using Size Exclusion Chromatography including, but not limited to, sepharose S
100, S200, 5300, superdex resin columns and their equivalents and comparables.

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.
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 sepharose FF column, SP Sepharose~ FF column, Q
Sepharose High Performance Column, Blue Sepharose FF column , Blue Column, Phenyl Sepharose FF column, DEAE Sepharose FF, or Methyl Column.
Additionally, albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO 00/44.772 which is herein incorporated by reference .in its entirety. One of skill in the art could easily modify the process described therein for use in the purification of albumin fusion proteins of the 1 S invention.
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 eukaryotic 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 inodifed 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 2S ~ 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.
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, 3S to the relatively low affinity of alcohol oxidase for OZ. Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol. oxidase genes (AOXI ) is highly active. In the presence of methanol, alcohol oxidase produced from the AOXI , gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris. See Ellis, S.B., et al., Mol. Cell. Biol.
S:I11I-2I
(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 h-anscriptional regulation of all or part of the ADXI regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
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 AOXI promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secrefory signal peptide (i.e., leader) located upstream of a multiple cloning site.
Many other yeast vectors could be used in place of pPIC9K, such ~ ~as-, pYES2, pYDI, pTEFl/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. .
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.
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, Nata~re, 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 ai-a not limited to, to the D-isomers of the common amino acids,, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline', sarcosine, citrulline,.homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglyeine, 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).
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 earned out by known techniques, including but' not limited, to specific chemical-cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH~; acetylation, formylation, oxidation, reduction;
metabolic synthesis in the presence. of tunicamycin; etc. r , Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-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 O-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.
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, fluoresceim 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 ('z'I,'zsl~ ~zsl~ 131I)~ carbon ('4C), sulfur (3sS),'tritium (3H), indium ('1'In, llzln, . ~113mIn' Ilsmln), technetiurri (99Tc,99mTc), thallium (z°1Ti), gallium (68Ga, 6'Ga), palladium (lospd), molybdenum (99Mo), xenon ('33Xe), .fluorine (18F), is3Sm, "'Lu, 's9Gd, '49Pm, l4oLa~ msl,b~ 166Ho~ 9oY~ a~Sc~ ls6Re~ ,ssRe~ t4zPr~ losRh~ and 9'Ru.
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, "'Lu, 9°Y, 166Ho, and ls3Sm, to polypeptides.
In a preferred embodiment, the radiometal ion associated with the ~macrocyclic chelators is '1'In. 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,N',N",N"'-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 m 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); which are hereby incorporated by reference in their entirety.
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 v 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 arginyl~tion, and ubiquitination. (See, for instance, PROTEINS - STRUCTURE AND MOLECULAR
r 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 (QrAGEN, 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 th'e "flag" tag. , 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, dibrorriomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum , (II) (DDP) cisplatin), antliracyclines (e.g., daunorubicin (formerly ~ daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic' agents (e:g., vineristine and vinblastine)~.
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-interferbti, 13-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 97133899); AIM II (See; International Publication No. WO 97/34911), Fas 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 ("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. Techniques for conjugating such therapeutic moiety to proteins (e.g., albumin fusion proteins) are well known in the art.
Albumin fusion proteins may also be attached to solid supports, which are particularly useful for immunoassays ox 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.
Albumin fusion proteins, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factors) and/or cytokine(s) can be used as a therapeutic.
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.
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 VH
protein will associate (either covalently or non-covalently) post-translationally.
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 (VH/VL,) pairs of the original antibody that binds a Therapeutic protein and the scFv fused to the C-terminus yof albumin would correspond to the other heavy/light (VH/VL) pair of the original antibody that binds a Therapeutic protein.
Also pi:ovided 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 immunogenicity (see U.S. Patent No. 4,179,337).' The chemical moieties for derivitization maybe selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the tike.
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.
The polymer may be of any molecular weight, and may be branched or unbranched.
For polyethylene glycol, the preferred molecular weight is between about l 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 WO 01/79442 ' PCT/USO1/11850 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, 12500, 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 kDa.
As noted above, the polyethylene glycol may have a branched structure.
Branched , polyethylene~gIycols axe described, for example, in U.S. Patent N.o.
5,643,575; Morpurgo et al., Appl. Biochem. Biotechhol. ' 56:59-72 (1996); Vorobjev et al., Nucleosides Nucleotides 18:2745-2750 (1999);~J and Caliceti et al., Biocoajug. Chem.
10:638-646 (1999), the disclosures.of each of which are incorporated herein by reference.
The polyethylene glycol molecules (or other chemical moieties) should be attached to the protein with consideration of effects on functional 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), herein incorporated by reference; see also Malik et al., Exp. H~ematol. 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 glutarriic, 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.
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 r cystei~ne) 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.
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 pegylate.d 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 10. 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.
As indicated above, pegylation of the albumin fusion proteins of the invention may be accomplished by. any number of means. Fox 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, the disclosures of each of which are incorporated herein by reference.
One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (C1SOZCHZCF3). 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.
Polyethylene glycol-can also be attached to proteins using a number of ,different intervening linkers. For example, U.S. Patent No. 5,612,460, the entire disclosure of which is incorporated herein by reference, 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-succinimidylsuccinate, MPEG activated with l,l'-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 axe described in International Publication No. W0~98/32466, the entire disclosure of which is incorporated herein by reference. Pegylated protein products produced using the reaction chemistries set out herein are included within the scope of the invention.
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 maybe linked, on average, to,I, 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).
The polypeptides of the invention .can be recovered and purified from chemical synthesis and recombinant cell cultures by standard irzethods 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.
. 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 of the Polynucleoti~des 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.
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 ., , ~ 137 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.
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 17 and 18).
Uses of the Polypeptides ..
' Each of the polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
Albumin fusion proteins of the invention are useful to provide immunological probes for differential identification of the tissues) (e.g., immunohistochemistry assays such as, for example, AB.C immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29:577-, (1981)) or cell type.(s) (e.g., immunocytochemistry assays).
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 2S 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 (13~I, ~zsl~
123I~ 1211), carbon (14C), sulfur (355), .tritium (3H), indium (1'S'"In, 113mIn, 112In, 'llln), and technetium (99Tc, 99mTC), thallium (2°'Ti), gallium (68Ga, 6'Ga), pallad.iuri~ (losPd), molybdenum (99Mo), xenon (133Xre) ~uorine (~sF') ~sssm m~Lu ~s9Gd m9Pm moLa' msYb 166Ho 9o.Y ~~Sc > > > > > > > > > > >
' ' 's6Re, '$$Re,'4~Pr,. '°SRh, 9'Ru; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin. ' Albumin fusion 'proteins of the invention can also be detected i~ vivo by imaging.
Labels or markers for ire vivo imaging of protein include those detectable by X-radiography, ~35 nuclear magnetic resonance (NMR) or electron spin relaxtion (ESR). For X-radiography, suitable labehs 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.
An albumin fusion protein which has been labeled with an appropriate detectable imaging moiety, such as. a radioisotope (for example, 13'I, 1'zln, 99mTc~
(~s~I~ ~zsl~ ~zsl, ~z~I)~
carbon ('4C), sulfur (35S), tritium (3H), indium (llsmln, 'l3mln,ulzTn, 111In), and technetium (9~c~ 99mTC), thallium (z°'Ti), gallium (68Ga, 6'Ga), palladium (losPd), molybdenum (99Mo), xenon. (133Xe), Buorine (isF, is3Sml i~~Lu, is9Gd~ i49Pm~ i4oLa~
ms~,b~ 166Ho~ soy 4'Se, 186Re, '$$Re, '4zPr, '°sRh, 9'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, e~ctracellular 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 wivo W mor .
imaging is described in S.W. Burchiel et al., "Immimopharmacokinetics 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.
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.
L _ 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.
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, 21381, or other radioisotopes such as, for example,'°3Pd, lssXe '3'I, 6sGe, 5'Co, 6sZn, ssSr, 32P, 3sS~ 90~,~
issSm~ issGd~ 169~yb~ slCr, ~Mn, 'sSe, 1135n; 9°Yttrium, '1'Tin, 's6Rhenium, '66liolmium, and lssRhenium; 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 9°Y. 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, "iIn. 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 '3'I.
Techniques known in the art may be applied to label polypepti.des of the invention.
Such techniques include, but are not limited to, the use of bifunctional conjugating agents (see e.g., U.S. Patent Nos. 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; the contents of each of which are hereby incorporated by reference in its entirety).
The albumin fusion proteins of the present invention are useful for diagnosis, treatment, prevention andlor 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.
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.
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).
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 sacpra, 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 polypeptidee to which the Therapeutic antibody used to make the albumin fusion protein immunospecifically binds, and/or reduce overproduction of the polypeptide to which the Therapeutic antibody ' used , , to make the albumin fusion protein immunospecifically binds. ~
Similarly, administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody. cari activate the polypeptide to which the Therapeutic antibody used. to make the albumin fusion protein immunospecifically binds, by binding to the polypeptide bound to.a membrane (receptor). . .
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 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.
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 levels) compared to the standard is indicative of a disorder. These diagnostic assays may be performed in vivo or ih vitro, such as, for example, on blood samples, biopsy tissue or autopsy tissue.
The present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome By "assaying the expression level of the gene encoding a polypeptide" is intended qualitatively or quantitatively measuring or estimating the Ieve1 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.
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.
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 Chomczynski 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, S 1 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).
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 polypeptides. 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 methbd.
Assaying polypepdde 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 radioisotopes, such as iodine ('zSI, 'z'I), carbon ('4C), sulfur (35S), tritium (3H), indium ("zIn), and technetium (99"'Tc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
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 IO and Lane (Harlow, E. and Lane, D., 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York), which is incorporated herein by reference in its entirety. 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 im 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. ' 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.
In a preferred embodiment, albumin fusion proteins comprising at least a fragment or variant of an antibody that immunospecifically binds at least a Therapeutic protein disclosed herein (e.g., the Therapeutic proteins disclosed in Table 1) ar 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 cytometric, or fluorimetric detection. ~ ' The albumin fusion proteins of the present invention may, additionally, be employed histologically, as in immunofluorescence, immunoelectron 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.
Immunoassays and non-immunoassays that detect polypeptides that bind to, are bound by, or associate with albumin fusion proteins will typically comprise incubating a S 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.
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.
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 un 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.
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 experimentati on. , .
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.
Labels or markers for ire 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.
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 ire vitro diagnostic procedures.
A polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example;
'3'I, "zIn, ~''"Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is 1 S 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 99mTc. 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 l3 in Tumorlmaging: The Radiochemical Detection of Cancer, S.W.
Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
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 Immunosorbent 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. Ejzzymol. 73:482-523 (1981);~Maggio, E. (ed.), 1980,.Enzyme Immunoassay, CRC Press, Boca Raton, FL" Ishikawa, E. et al., (eds.), 1981, Enzyme Immunoassay, Kgaku 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 chein.ical 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 oxidase, 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.
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 Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference herein). The radioactive isotope can be detected by means including, but not limited to, a gamma counter, a scintillation counter, or autoradiography.
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.
The albumin fusion protein can also lie detectably labeled using fluorescence emitting metals such as 's2Eu, 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).
~ The albumin fusion proteins can also can be detectably labeled by coupling it to a chemiluminescenf compound. The presence of the chemiluminescent-tagged albumin fusion protein is then deternnined 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, theramatic acridinium ester, imidazole, acridinium salt and oxalate ester.
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.

Trans~:enic Organisms 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 . 5 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 IO 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.
1 S ' 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 information 20 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.
A transgenic organism may be a transgenic animal or a transgenic plant.
Transgenic 25. ~ 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 30 Biology No. 62, Humana Press (1997)): The method of introduction of nucleic acid fragments into recombination competerit mammalian cells can be by any method which favors co-transformation of multiple nucleic acid molecules. Detailed procedures for producing transgeriic 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.
35 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 148 , regulatory factor 1 (IRF-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.5. Patent No.
5,720,936); have a reduced capacity fo mediate cellular adhesion (U.S. Patent No. 5,602,307);
possess a bovine growth hormone gene (Clutter, et al. (1996) Genetics I43(4):1753-1760);
or, are capable of generating a fully human antibody response (McCarthy ( 1997) The Lancet 349(9049):405).
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):609-6I7;, 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).
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 lactalbumin (see, e.g., DiTullio (1992). BioTechnology I0:74-77;
Clark et al.
(1989) BioTechnology 7:487-492; Gorton et al. (1987) BioTechnology 5:1183-1287; 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.
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 (e.g., see Example 19). 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, all of which are hereby incorporated by reference.
Pharmaceutical or Therapeutic Compositions The albumin fusion proteins of the invention or formulations thereof may be . 149 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.
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 carriers) 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 maybe formulated well in advance in aqueous form, for instance, weeks or months or longer time periods before being dispensed.
For example, wherein the Therapeutic protein is hGH, EPO, alpha-IFN or beta-IFN, 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 exhibited in Table 2, most ,Therapeutic proteins are unstable with short shelf-lives after formulation with an aqueous carrier. As discussed above, the shelf-life of many of these Therapeutic proteins are markedly increased or prolonged after fusion to HA.
Table 2 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 o~ 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.
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.
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.
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-agueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or mufti-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.
As an example, when an albumin fusion protein of the invention comprises growth hormone 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 albunnin fusion proteins compared to that of native hGH. Growth hormone is typically administexed at 0.3 to 30.0 IUlkg/week~ for example 0.9 to 12.0 IU/kg/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, fox example to twice a week, once a week or less.
Formulations or compositions of the invention rmay be packaged together with, or included in a k_it 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 coilditions, 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.
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 -15 Hegenhart, Food Product Design, Dec. 1993.. ' 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.
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.
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/hpur, 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.
Albumin fusion proteins andlor polynucleotides can be ire 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, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion. .
Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustairied-release systems. Examples of sustained-release albumin fusion proteins and/or polynucleotides are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as liy 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 type. I~ 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 andlor 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).
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). - . ' .
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 Liposomes 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. Appl. 83-118008; ~U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP
102,324. Ordinarily, the liposomes 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.

WO 01/79442 ~ PCT/USO1/11850 proportion being adjusted for the optimal Therapeutic.
In yet an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are delivered by way of a pump (see Langei-, 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)).
Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
For parenteral administration, in one embodiment, the albumin fusion protein andlor 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.
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.
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, succinate, acetic acid,Jand other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins;
hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; nionosaccharides, disaccharides, and other carbohydrates 30- including cellulose or its derivatives, glucose, tnanose,. or dextrins;
chelating agents such as EDTA; sugar. alcohols such as mannitol or sorbitol; counterions such as sodium; and/or _ nonionic surfactants such as polysorbates~ poloxamers, br PEG.
The albumin fusion protein is typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH of about 3 to 8. It will be understood that the use of-certain of the foregoing excipients, carriers; or stabilizers will .result in the formation of polypeptide .salts. .
Any pharmaceutical used for therapeutic administration can be sterile.
Sterility is readily accomplished by filtration through sterile filtration .membranes (e.g., 0.2 micron membranes).. Albumin fusion proteins and/or polynucleotides generally are placed into a container having a sterile access port, for example, an intravenous solution bag nor vial having a stopper pierceable by a hypodermic injection needle.
Albumin fusion proteins and/or polynucleotides ordinarily will be stored in unit or mufti-dose containers, for example, sealed 'ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. As an example of a lyophilized formulation, 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous albumin fusion protein andlor polynucleotide solution, and the resulting mixture is lyophilized. The infusion solution is prepared by reconstituting the lyophilized albumin fusion protein and/or polynucleotide using bacteriostatic Water-for-Injection.
In a specific and preferred embodiment, the Albumin fusion protein formulations comprises 0.0I M sodium phosphate, 0.15 mM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2. In another specific and preferred embodiment, the Albumin fusion protein formulations consists 0.01 M sodium phosphate, 0.15 mM sodium chloride, O.I6 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2. The pH and buffer are chosen to match physiological conditions and the salt is added as a tonicifier. Sodium octanoate has been chosen due to its reported ability to increase the thermal stability of the protein in solution. Finally, polysorbate has been added as a generic surfactant, which lowers the surface tension of the solution and lowers non-specific .
adsorption of the albumin fusion protein to the container closure system.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the albumin fusion proteins and/or polynucleotides of the invention. Associated with such containers) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In addition, the albumin fusion proteins and/or polynucleotides may be employed in conjunction with other therapeutic compounds.
The albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in,combination with. adjuvants. Adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, alum, alum plus deoxycholate (ImmunoAg), MTP-PE (Biocine Corp.), (Genentech, Inc.), BCG (e.g., THERACYS~), MPL and nonviable preparations of Corynebacterium parvum. In a specific embodiment, albumin fusion proteins and/or polynucleotides' of the invention are administered- in combination with alum.
In another specific embodiment, albumin.fusion proteins and/or polynucleotides of the invention are WO 01/79442 . PCT/USO1/11850 administered in combination with QS-21. Further adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, lVlonophosphoryl .lipid immunomodulator, AdjuVax 100a, QS-21~, QS-18, CRL;1005, Aluminum salts, MF-59, and Virosomal adjuvant technology. Vaccines that may be administered with the albumin fusion proteins ,and/or polynucleotides of the invention include, but are not limited to, vaccines directed toward protection against MMR
(measles, mumps, rubella), polio, varicella, tetanus/diptheria, hepatitis A, hepatitis B, Haemophilus influenzae B, whooping cough, pneumonia, influenza, Lyme's Disease, ' rotavirus, cholera, yellow fever, Japanese encephalitis, poliomyelitis, rabies, typhoid fever, and . pertussis: Combinations may be administered either concomitantly, e.g., as. an admixture, separately but simultaneously or concurrently; or sequentially.
This includes presentations in which the combined agents are administered together as a therapeutic mixture, and also procedures in which the combined agents are administered separately but simultaneously, e.g., as through separate intravenous lines into the same individual.
Administration "in combination" further includes the separate administration of one of the compounds or agents given first, followed by the second.
The albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with other therapeutic agents. Albumin fusion protein and/or polynucleotide agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include but not limited to, chemotherapeutic agents, antibiotics, steroidal and non-steroidal' anti-inflammatories, conventional immunotherapeutic agents, and/or therapeutic treatments described below.
Combinations may be administered either concomitantly, e.g., as an admixture,' separately but simultaneously or concurrently; or sequentially. . This includes presentations in which the combined agents are administered together as a therapeutic mixture, and also procedures in which the combined agents are administered separately but simultaneously, e.g.,. as through separate intravenous Lines into the same. individual. Administration "in combination" further includes the separate administi-ation~ of one of the compounds or agents given first, followed by the second.
. In one~~ embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an anticoagulant.
Anticoagulants that may be administered with the compositions of the invention include, but are not limited to, heparin, low molecular weight heparin, warfarin sodium (e.g.; COUMADIN~), dicumarol, 4-hydroxycoumarin, anisi.ndione (e.g., MIRADONT"'), acenocoumarol (e.g., nicoumalone, SINTHROMET"'), indan-1,3-dione, phenprocoumon (e.g., . MARCUMARTM), ethyl biscoumacetate (e.g., TROMEXANTM), and aspirin: In a specific embodiment, compositions of the invention are administered in combination with heparin and/or warfarin.

In another specific embodiment, compositions of the invention are administered in combination with warfarin. In another specific embodiment, compositions of the invention are administered in combination with warfarin and aspirin. In another specific embodiment, compositions of the invention are administered in combination with heparin. In another specific embodiment, compositions of the, invention are administered in combination with heparin and aspirin.
In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with thrombolytic drugs.
Thrombolytic drugs that may be administered with the compositions of the invention include, but are not limited to, plasminogen, lys-plasminogen, alpha2-antiplasmin, streptokinae (e.g., KABIKINASETM), antiresplace (e.g., EMINASETM), tissue plasminogen activator (t-PA, altevase, ACTIVASET"'), urokinase (e.g., ABBOKINASETM), sauruplase, (Prourokinase, single chain urokinase), and aminocaproic acid (e.g., AMICART"'). In a specific embodiment, compositions' of the invention are administered in combination with tissue plasminogen activator and aspirin.
In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiplatelet drugs.
Antiplatelet drugs that may be administered with the compositions of the invention include, but are not limited to, aspirin, dipyridamole (e.g., PERSANTINETM ), and ticlopidine (e.g., TICLIDTM
).
In specific embodiments, the use of anti-coagulants, thrombolytic and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for. the prevention, diagnosis, and/or treatment of thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina: In specific embodiments, ~ the use of anticoagulants, thrombolytic drugs and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention of ~ occulsion of saphenous grafts, for reducing the risk of periprocedural thrombosis as~ might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonxheumati,c atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mitral valves disease. Other uses for the therapeutics of the invention, alone or in combination with antiplatelet, anticoagulant, and/or throrribolytic drugs, include, but are not limited to, the prevention of occlusions in extracorporeal devices (e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines).
In certain embodiments, albumin fusion 'proteins and/or polynucleotides of the invention are administered in combination with anti~retroviral agents,.
nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and/or protease inhibitors (PIs). NRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, RETROVIR( (zidovudine/AZT), VIDEX( (didanosine/ddI), HIVID( (zalcitabine/ddC), ZERIT( (stavudine/d4T), EPIVIR( (lamivudine/3TC), and COMB1VIR( (zidovudine/lamivudine). NNRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, VIRAMUNE( (nevirapine), RESCRIPTOR( (delavirdine); and SUSTIVA( (efavirenz). Protease inhibitors that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, CRIXIVAN( (indinavir), NORVIR( (ritonavir), INVIRASE( (saquinavir),.and VIRACEPT( (nelfinavir). In a specific embodiment, _antiretroviral agents, nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors, and/or protease inhibitors may be used in ' any combination with albumin fusion proteins andlor polynucleotides of the invention to treat AIDS and/or to prevent or treat HIV
infection.
Additional NRTIs include LODENOSINE( (F-ddA'; an acid-stable adenosine NRTI; Triangle/Abbott; COVIRACIL( (emtricitabine/FTC;
structurally related to lamivudine (3TC) but with 3- to 10-fold greater activity in vitro; Triangle/Abbott); dOTC (BCH-10652, also structurally related to lamivudine but retains activity against a substantial proportion o f 20~ lamivudine-resistant isolates; Biochem Pharma); Adefovir (refused.
approval for ,anti-HIV therapy by FDA; Gilead Sciences); PREVEON( (Adefovir Dipivoxil, the active prodrug of adefovir; its active form is PMEA-pp);
TENOFOVIR( (bis-POC PMPA, a ,PMPA prodrug; Gilead); DAPD%DXG
(active metabolite of DAPD; Triangle/Abbott); D-D4FC (related to 3TC, with activity against ' AZT/3TC-resistant virus); GW420867X (Glaxo Wellcome);
ZIAGEN( (abacavir/159IJ89; Glaxo Wellcome Inc.); CS-87 (3'azido-2',3'-dideoxyuridine; WO 99/66936); and S-acyl-2-thioethyl (SATE)-bearing prodrug forms of (-L-FD4C and (-L-FddC (WO 98/17281).
Additional NNRTIs include COACTINONT"' (Emivirine/MKC-442, potent NNRTI
of the HEFT class; Triangle/Abbott); CAPRAVIRINET"' (AG-1549/S-1153, a next generation NNRTI with activity against viruses containing the K103N mutation;
Agouron);
PNU-142721 (has 20- to 50-fold greater activity than its predecessor delavirdine and is active against K103N mutants;. Pharmacia & Upjohn); DPC-961 and DPC-963 (second-generation derivatives of efavirenz, designed to be active against viruses with the K103N
mutation; DuPont); GW-420867X (has 25-fold greater activity than HBY097 and is active against K103N mutants; Glaxo Wellcome); CALANOLIDE A (naturally occurring -agent from the latex tree; active against viruses containing either or both the Y
181C and K103N
mutations); and Propolis (WO 99149.830).
Additional protease inhibitors include LOPINAVIRT"" (ABT378/r; Abbott Laboratories); BMS-232632 (an azapeptide; Bristol-Myres Squibb); .
TIPRANAVIRT"' . (PNU-140690, a non-peptic dihydropyrone; Pharmacia & Upjohn); PD-178390 (a nonpeptidic. dihydropyrone; Parke-Davis); BMS 232632 (an azapeptide; Bristol-Myers Squibb); L-756,423 (an indinavir, analog; Merck); DMP-4.5'0 (a cyclic urea compound; Avid & DuPont); AG-1776 (a peptidomimetic with irz vitro activity against protease inhibitor-resistant viruses; Agouron); VX-175/GW-433908 (phosphate prodrug of amprenavir;
Vertex & Glaxo Welcome); CGP61755 (Ciba); and AGENERASET"" (amprenavir; Glaxo Wellcome Inc.).
Additional antiretroviral agents include ~ fusion' inhibitors/gp4l .binders.
Fusion inhibitors/gp41 binders include T-20 (a peptide from residues 643-678 of the HIV gp41 transmembrane protein ectodomain which binds to gp41 in its resting state and prevents ,15 transformation to the fusogenic state; Trimeris) and T-1249 (a second-generation fusion inhibitor; Trimeris).
Additional antiretroviral agents include - fusion inhibitors/chemokine receptor antagonists. Fusion inhibifors/chemokine receptor antagonists include CXCR4 antagonists such as AMD 3100 (a bicyclam), SDF-1 and its analogs, and ALX40-4C (a cationic peptide), T22 (an 18 amino acid peptide; Trimeris) and'the T22 analogs T134 and T140;
CCRS antagonists such as RANTES (9-68), AOP-RANTES, NNY-RANTES, and TAK-779; and CCRSICXCR4 antagonists- such as. NSC 651016 (a distamycin analog).
Also included are CCR2B, CCR3, and CCR6 antagonists. Chemokine recpetor agonists such as RANTES, SDF-1, MIP-la, MIP-1(3, etc., may also inhibit fusion.
Additional, antiretroviral agents include integrase inhibitors. Integrase inhibitors include dicaffeoylquinic (DFQA) acids; L-chicoric acid (a dicaffeoyltartaric (DCTA) acid);
quinalizarin (QLC) and related anthraquinones; ZINTEVIRT"' (AR 177, an oligonucleotide that probably acts at cell surface rather than being a true integrase in$ibitor; Arondex); and . naphthols such as, those disclosed in WO 98150347. ~ ' , Additional antiretroviral agents include hydroxyurea-like compunds such as (a purine nucleoside phosphorylase inhibitor; Biocryst); ribonucleotide reductase inhibitors such as DIDOXT"" (Molecules for Health); inosine monophosphate dehydrogenase (IMPDH) inhibitors sucha as VX-497 (Vertex); and mycopholic acids such as CellCept (mycophenolate mofetil; Roche).

Additional antiretroviral agents include inhibitors of viral integrase, inhibitors of viral genome nuclear translocation such as arylene bis(methylketone) compounds; inhibitors .z of HIV entry such as AOP-RANTES, NNY-RANTES, RANTES-IgG fusion protein, soluble complexes of RANTES and glycosaminoglycans (GAG), and AMD-3100;
nucleocapsid zinc finger inhibitors such as dithiane compounds; targets of HIV
Tat and Rev;
and pharrriacoenhancers such as ABT-378.
Other antiretroviral therapies and adjunct therapies include cytokines and-lymphokines such as MIP-la, MIP-1(3, SDF-la, IL-2, PROLEUKINT""
(aldesleukin/L2-7001; Chiron), IL-4, IL-10, IL-12, and IL-13; interferons such as IFN-a2a;
antagonists of TNFs, NFtcB, GM-CSF, M-CSF, and IL-10; agents that modulate immune activation such as cyclosporin and prednisone; vaccines such as RemuneT"' (HIV Immunogen)., 003 (Apollon), recombinant gpI20 and fragments, bivalent (B/E) recombinant envelope glycoprotein, rgpI20CM235, MN rgp120, SF-2 rgp120, gp120/soluble CD4 complex, Delta JR-FL protein, branched synthetic peptide derived from discontinuous gp120 C3/C4 ~ domain, fusion-competent immunogens, and Gag, Pol, Nef, and Tat vaccines;
gene-based therapies such as genetic suppressor elements (GSEs; WO 98/54366), and intrakines (genetically modified CC chemokines targetted to the ER to block surface expression of newly synthesized CCRS (Yang et al., PNAS 94:11567-72 (1997); Chen et al., Nat. Med.
3:1110-16 (1997)); antibodies such as the anti-CXCR4 antibody .1265, the anti-CCRS
antibodies 2D7, 5C7, PAB, PA9; PA10, PA11, PA12, and PA14, the anti-CD4 antibodies Q4120 and RPA-T4, the anti-CCR3 antibody 7B11., the anti-gp120 antibodies 17b, 48d, 447-52D, 2S7-D, 268-D and 50.1, anti-Tat antibodies, anti-TNF-a antibodies, and monoclonal, antibody 33A; aryl hydrocarbon (AH) receptor agonists and antagonists such as TODD, ' 3,3',4,4',5-pentachlorobiphenyl, 3,3',4,4'-tetrachlorobiphenyl, and a-2S naphthoflavone (WO 98/30213); and antioxidants such as y-L-glutamyl-L-cysteine ethyl ester (y-GCE; WO 99/56764).
In. a 'further embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiviral agent.. Antiviral agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, acyclovir, ribavirin, amantadine, and remantidine.
In other embodiments, albumin fusion proteins and/or polynucleotides of the invention may be administered in combination with anti-opportunistic infection agents.
Anti-opportunistic agents that may be administered in combination with the albumin fusion proteins andlor polynucleotides of the invention, include, but are not limited to, TRIMETHOPRIM-SULFAMETHOXAZOLET"', DAPSONET"", , PENTAMIDINET"', ATOVAQUONET"', ISONIAZIDT"", RIFAMPINT"', PYRAZINAMIDET"", ETHAMBUTOLT"', RIFABUTINT"', CLARITHROMYCINT"', AZITHROMYCINT"", GANCICLOVIRT"", FOSCARNETT"", CIDOFOVIRT"", FLUCONAZOLET"', ITRACONAZOLET"', I~ETOCONAZOI:ET"", . ACYCLOVIRT'", FAMCICOLVIRT"', PYRIMETHAMINET"', LEUCOVORINT"", NEUPOGENT"' (filgrastim/G-CSF), and LEUKINET"" (sargramostim/GM-CSF). In a specific embodiment,.albumin fusion proteins and/or polynucleotides of the invention are used in any combination with TRIMETHOPRIM-SULFAMETHOXAZOLET"', DAPSONET"", PENTAMIDINET"", and/or ATOVAQUONET"' to prophylactieally treat or prevent an opportunistic P~eeumocystis carinii pneumonia infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ISONIAZIDT"", RIFAMPINT"', PYRAZINAMIDET"', and/or ETHAMBUTOLT"' to prophylactically treat or .
prevent an opportunistic Mycobacterium avium complex infection. In another specific ~ embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with RIFABUTINT"', CLARITHROMYCINT"', and/or AZITHROMYCINT"' to prophylactically treat or prevent an opportunistic .Mycobacterium tuberculosis infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the ,invention are used in any combination with GANCICLOVIRT"', FOSCARNETT"', and/or CIDOFOVIRT"' to prophylactically treat or prevent an opportunistic cytomegalovirus infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with FLUCONAZOLET"', ITRACONAZOLEr"", and/or KEfOCONAZOLET"' to prophylactically treat or prevent an opportunistic fungal infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ACYCLOVIRT"' and/or FAMCICOLVIRT"' to prophylactically treat or prevent an opportunistic herpes simplex virus type I and/or type II infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with .
PYRIMETHAMINET"' and/or LEUCOVORINT"' to prophylactically treat or prevent an opportunistic Toxoplasma gondii infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention , are used in any combination with LEUCOVORINT"' and/or NEUPOGENT"' to prophylactically treat or prevent an opportunistic bacterial infection.
Iri a further embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antibiotic agent. Antibiotic agents that inay be administered with the albumin fusion proteins andlor polynucleotides of the invention include, but are not limited to, amoxicillin, beta-lactamases, aminoglycosides, beta-lactam (glycopeptide), beta-lactamases, Clindamycin, chloramphenicol, cephalosporins, ciprofloxacin, erythromycin, fluoroquinolones, macrolides, metronidazole, penicillins, quinolones, rapamycin, rifampin, streptomycin, sulfonamide, tetracyclines, trimethoprim, trimethoprim-sulfamethoxazole, and vapcomycin. .
In other embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunestimulants.
Immunostimulants that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to; levamisole (e.g., ERGAMISOLTM), isoprinosine (e.g. INOSIPLEXTM), interferor;s (e.g. interferon alpha), and interleukins .(e.g., IL-2).
' In other embodiments, albumin fusion proteins and/or ~ polynucleotides ~of the - invention are administered in combination with immunosuppressive ' agents.
Immunosuppressive agents that may be administered in combination with 'the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, steroids, cyclosporine, cyclosporiile analogs, cyclophosphamide methylprednisor<e, prednisone, azathioprine, FK-506, 15-deoxyspergualin, and other immunasuppressive agents that act by suppressing the function of responding T cells. Other immunosuppressive agents that mzay be administered in combination with the albumin fusion proteins and/or polynucleotides of the ~ invention include, but are not limited to, prednisolone, methotrexate, thalidomide, methoxsalen, rapamycin, leflunomide, mizoribine (BREDININTM), brequinar, deoxyspergualin, and azaspirane (SKF. 105685), ORTHOCLONE OKT~ 3 (muromonab-CD3), SANDIMMUNET"', NEORALT"', SANGDYAT"' (cyclosporine), PROGRAF~ (FK506, tacrolimus), CELLCEPT~
(mycophenolate motefil, of which the active metabolite is mycophenolic acid), IMURANTr'' (azathioprine), glucocorticosteroids, adrenocortical , steroids such as DELTASONETM
(prednisone) and HYDELTRASOLT"' ~ (prednisolone), FOLEXTM and MEXATET'"
(methotrxate), OXSORALEN-ULTRATM (methoxsalen) and RAPAMUNET"' (sirolimus).
In a specific embodiment, immurlosuppressants may be used to ,prevent rejection of organ or 162' bone marrow transplantation.
In an additional embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered alone or in combination with one or more intravenous immune globulin preparations. Intravenous immune globulin preparations that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but not limited to, GAMMART"", IVEEGAMT"', SANDOGLOBULINT"", GAMMAGARD SIDT"', ATGAMT"' (antithymocyte glubulin), and GAMIMUNEr'". In a specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with intravenous immune globulin preparations in transplantation therapy (e.g., bone marrow transplant).
In certain embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered alone or.in combination with an anti-inflammatory agent. Anti-inflammatory agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, corticosteroids (e.g.
betamethasone, budesonide, cortisone; dexamethasone, hydrocortisone,' methylprednisolone, prednisolone, piednisone, and triamcinolone), nonsteroidal anti-inflammatory drugs (e.g., diclofenac, diflunisal, etodolac, fenoprofen, floctafenine, flurbiprofen, ibuprofen, indomethacin, ketoprofen, meclofenamate, meferiamic acid, meloxicam, nabumetone, naproxen; ' oxaprozin, phenylbutazone, piroxicam, sulindac, tenoxicam, tiaprofenic acid, and tolmetin.), as well as antihistamines, aminoarylcarboxylic acid derivatives, arylacetic acid derivatives,' arylbutyric acid derivatives, arylcarboxylic acids, arylpropionic acid derivatives, pyrazoles, ~ pyrazolones, salicylic acid derivatives, thiazinecarboxamides, e-acetamidocaproic acid, ,S-adenosylmethionine, 3-amino-hydroxybutyric acid, amixetrine, . bendazac, benzydamine, bucolome, difenpiramide, di,tazol, emorfazone, guaiazulene, nabumetone, nimesulide, orgotein, oxaceprol,, paranyline, 'perisoxal, pifoxime, proquazone, proxazole, and tenidap.
In an additional embodiment, the compositions of the invention are administered alone or in combination with an anti-angiogenic agent. Anti-angiogenic agents that may be administered with the compositions of the invention include, but are not limited to, Angiostatin (Entremed, Rockville, MD), Tro~onin-1 (Boston Life Sciences, Boston, MA), anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel (Taxol), Suramin, Tissue Inhibitor of Metalloproteinase-l, Tissue Inhibitor of Metalloproteinase-2, VEGI, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter "d group" transition metals.
Lighter "d group" transition metals include, for example; vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition.metal species may form transition metal complexes_ Suitable complexes of the above-mentioned transition.metal species include oxo transition metal complexes. ~ -Representati.ve~ examples of vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes. Suitable vanadate complexes include metavanadate and orthovanadate complexes such as, for example, ammonium metavanadate, sodium metavanadate, and sodium orthovanadate: Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates.
Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes. Suitable tungstate complexes include ammonium tungstate, calcium tungstate, sodium tungstate dihydrate, and tungstic acid. Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) oxide. Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates. Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum (VI) oxide, and molybdic acid. Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate. Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars.
A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include, but are not limited to;
platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res. 51:22-26, (1991)); Sulphated Polysaccharide Peptidoglycan Complex (SP- PG) (the function of this compound may be enhanced by the presence of steroids such as estrogen, and tamoxifen citrate); Staurosporine;
modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fi~marate; 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin;
Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. Bio. Chem. 267:I7321-17326, (1992));
Chymostatin (Tomkinson et al., Biochem J. 286:475-480, (1992)); Cyclodextrin Tetradecasulfate; Eponemycin; Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, (1990)); Gold Sodium Thiomalate ("GST"; Matsubara and Ziff, J. Clin.
Invest.
79:1440-1446, (1987)); anticollagenase-serum; alpha2-antiplasmin (Holmes et al., J. Biol.
Chem. 262(4):1659-1664, (1987)); Bisantrene (National Cancer Institute);
Lobenzarit disodium (N-(2)-carboxyphenyl-4- chloroanthronilic acid disodium or "CCA";
(Takeuchi et al., Agents Actions 36:312-316, (1992)); and metalloproteinase inhibitors such as BB94.
Additional anti-angiogenic factors that may also be utilized within the context of the WO 01/79442 ~ PCT/USO1/11850 present.invention include Thalidomide, (Celgene, Warren, NJ); Angiostatic steroid; AGM-1470 (H. Brem and J. Folkman J Pediatr. Sung. 28:445-51 (1993)); an integrin alpha v beta 3 antagonist (C. Storgard et al., J Clin. Invest. 103:47-54 (1999));
carboxyriaminolmidazole; Carboxyamidotriazole (CAI) (National Cancer Institute, Bethesda, MD); Conbretastatin A-4. (CA4P) (OXiGENE, Boston, MA); Squalamine (Magainin Pharmaceuticals, Plymouth Meeting, PA); TNP-470, (Tap Pharmaceuticals, Deerfield, IL); ZD-0101 AstraZeneca (London, UK); APRA (CT2584); Benefin, .
Byrostatin-1 (SC339555); CGP-41251 (PKC 412); CM101; Dexrazoxaile (ICRF187);
DMXAA; Endostatin; Flavopridiol; Genestein; GTE; ImmTher; Iressa (ZD1839);
Octreotide (Somatostatin); Panretin; Penacillamine; Photopoint; PI-88; Prinomastat (AG-3340) Purlytin; Suradista (FCE26644); Tamoxifen (Nolvadex); Tazarotene;
Tetrathiomolybdate;
Xeloda (Capecitabine); and 5-Fluorouracil.
Anti-angiogenic agents that may be administed in combination with the compounds of the invention may work through a variety of mechanisms including, but not limited to, inhibiting proteolysis of the extracellular matrix, blocking .the function of endothelial cell-extracellular matrix adhesion molecules, by antagonizing the function of angiogenesis inducers such as growth factors, and inhibiting integriri receptors expressed on proliferating endothelial cells. Examples of anti-angiogenic inhibitors that interfere with extracellular matrix proteolysis and which may be administered in combination with the compositons of the invention include, but are not lmited to, AG-3340 (Agouran, La Jolla, CA), 9566 (Bayer, West Haven, CT), BMS-275291 (Bristol Myers Squibb, Princeton, NJ), .
' . CGS-27032A (Novartis, East Hanover, NJ), Marimastat (British Biotech, Oxford, UK), and Metastat (Aeterna, St-Foy, Quebec). Examples of anti-angiogenic inhibitors that act by blocking the function of endothelial cell-extracellular matrix adhesion molecules and which may be administered in combination with the compositons of the invention include, but are not lmited to, EMD-121974 (Merck KcgaA Darmstadt, Germany) and Vitaxin (Ixsys, La Jolla, CA/Medimmune, Gaithersburg, MD). Examples of anti-angiogenic agents that act by directly antagonizing or inhibiting angiogenesis inducers ~ and which may be administered in combination with the compositons of the invention include, but are not Invited to, . 30 Arigiozyme (Ribozyme, Boulder, CO), Anti-VEGF antibody (Genentech, S. San Francisco, CA), PTK-787/ZK-225846 (Novartis, Basel, Switzerland), SU-101 (Sugen, S. San Francisco, CA), SU-5416 (Sugen/ Pharmacia Upjohn, Bridgewater, NJ); and SU-(Sugen). Other anti-angiogenic agents act to indirectly inhibit angiogenesis.
Examples of indirect inhibitors of angiogenesis which may be administered in combination with the , compositans of the invention include, but are not limited to, IM-862 (Cytran, Kirkland, WA), Interferon-alpha, IL-12 (Roche, Nutley, NJ), and Pentosan polysulfate (Georgetown University, Washington, DC).

In particular embodiments, the use of compositions of the invention in combination ve~ith anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of an autoimmune disease, such as for example, an autoimmune disease described herein.
In a particular embodiment, the use of compositions of,the invention in combination with anti-angioenic agents is contemplated for 'the treatment, prevention, andlor amelioration of arthritis. In ~a more particular embodiment, the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of rheumatoid arthritis. ' In another embodiment, the polynucleotides encoding a polypeptide of the present invention are administered in combination with an angiogenic protein, or polynucleotides encoding an angiogenic protein. Examples of angiogenic proteins that may be administered with the compositions of the invention include, but are not limited to, acidic and basic fibroblast growth factors; VEGF-1, VEGF-2, VEGF-3, epidermal growth factor alpha and beta, platelet-derived endothelial cell growth factor, platelet-derived growth factor, tumor necrosis factor alpha, hepatocyte growth factor, insulin-like growth factor, colony stimulating factor, macrophage . colony stimulating factor, granulocyte/macrophage colony stimulating factor, and nitric oxide synthase.
In additional embodiments, compositions of the invention are administered in combination with a chemotherapeutic agent. Chemotherapeutic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to alkylating agents such as nitrogen mustards (for example, Mechlorethamine, cyclophosphamide, Cyclophosphamide Ifosfami,de, Melphalan (L-sarcolysin), and Chlorambucil), ethylenimines and methylmelamines (for example, . Hexamethylmelamine and Thiotepa), alkyl sulfonates (for example, Busulfan), nitrosoureas .
(for example, Carmustine (BCNU), Lomustine (CCNU), Semustine (methyl-CCNU), and Streptozocin (streptozotocin)), triazenes (for example, Dacarbazine (DTIC;
dimethyltriazenoimidazolecarboxamide)), folic acid analogs (for example, Methotrexate (amethopterin)), pyrimidine analogs (for example, Fluorouacil (5-fluorouracil;
5-FU),,:
Floxuridine (fluorodeoxyuridine; FudR), and Cytarabine (cytosine arabinoside)), purine analogs and related inhibitors (for example, Mercaptopurine (6-mercaptopurine;
6-MP), ~Thioguanine (6-thioguanine; TG), and Pentostatin (2'-deoxycoformycin)), vinca alkaloids (for example, Vinblastine (VLB, vinblastine sulfate)) and Vincristine (vincristine sulfate)), epipodophyllotoxins (for example, Etoposide and Teniposide), antibiotics (for example, . Dactinomycin (actinomycin D), Daunorubicin (daunomycin; rubidomycin), Doxorubicin, Bleomycin, Plicamycin (mithramycin), and Mitomycin (mitomycin C), enzymes (for example, L-Asparaginase), biological response modifiers (for example, Interferon-alpha and interferon-alpha-2b), platinum coordination compounds (for example, Cisplatin (cis-DDP) and Carboplatin), anthracenedione (Mitoxantrone), substituted ureas (for example, Hydroxyurea), methylhydrazine ~ derivatives (for example, Procarbazine (N-methylhydrazine; MIH), adrenocorticosteroids (for example, Prednisone), progestins (for example, Hydroxyprogesterone caproate, Medroxyprogesterone, Medroxyprogesterone acetate, and Megestrol acetate), estrogens (for example, Diethylstilbestrol (DES), Diethylstilbestrol diphosphate, Estradiol, and Ethinyl estradiol), antiestrogens (for example, Tamoxifen), androgens .(Testosterone proprionate, and Fluoxymesterone), antiandrogens (for example, .Flutamide), gonadotropin-releasing horomone analogs (for example, Leuprolide), other hormones and hormone analogs (for example, methyltestosterone, estramustine, estramustine phosphate sodium, chlorotrianisene, and testolactone); and others (for example, dicarbazine, glutamic acid, and mitotane).
~In one embodiment, the compositions of the invention are administered in combination with one or more of the following drugs: infliximab (also known as ~15 RemicadeT"' Centocor, Inc.), Trocade (Roche, RO-32-3555), Leflunomide (also known as AravaTM from Hoechst Marion Roussel), KineretT'" (an IL-1 Receptor antagonist also known as Anakinra from Amgen, Inc.) .
In a specific embodiment, compositions of the .invention are administered in combination with CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) or combination of one or more of the components of CHOP. In one embodiment, the compositions of the invention are administered in combination with anti-CD20 antibodies, human monoclonal anti-CD20 antibodies. In another embodiment, the compositions of the invention are administered in combination with anti-CD20~ antibodies and CHOP, or anti-CD20 antibodies and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone. In a specific'embodiment, compositions of the invention are administered in combination with Rituximab. In a further embodiment, compositions of the invention are administered with Rituximab and CHOP, or Rituximab and any combination of one or more of the components of CHOP, particularly cyclophosphamide . and/or prednisone. In a specific embodiment, compositions of the invention are administered in combination with tositumomab. In a further embodiment, compositions of the invention are administered with tositumomab and CHOP, or tositumomab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone. The anti-CD20 antibodies may, optionally be associated with radioisotopes, toxins or cytotoxic prodrugs.
In another specific embodiment, the compositions of the invention are administered in combination ZevalinT"". _ In a further embodiment, compositions of the invention are administered with ZevalinT"' and CHOP, or ZevalinT"' and any combination of one or more 167 ' ' of the components of CHOP, particularly cyclophosphamide and/or prednisone.
ZevalinT""
may be associated with one or more radisotopes. Particularly preferred isotopes are 9°Y and mln~
In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with cytokines. Cytokines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, IL2, IL3, ILA~, ILS, IL6, IL7, IL10, IL12, IL13, IL15, anti-CD40, CD40L, IFN-gamma and TNF-alpha. In another embodiment, albumin fusion proteins and/or polynucleotides of the invention may be administered with any interleukin, including, but not limited to, IL-!alpha, IL-!beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21..
In one embodiment, the albumin fusion proteins and/or polynucleotides ~of the invention are administered in combination with members of the TNF_family. TNF, TNF-related or TNF-like molecules that may be administered with the albumin fusion . proteins and/or polynucleotides of the invention include, but are not limited to, soluble forms of TNF-alpha, lymphotoxin-alpha (LT-alpha, also known as TNF-beta), LT-beta (found ~in complex heterotrimer LT-alpha2-beta), OPGL, Fast, CD27L, CD30L, CD40L, 4-1BBL, DcR3, ,0~40L, TNF-gamma (International Publication No. WO 96/14328), AIM-I
(International Publication No. WO 97/33899), endokine-alpha (International Publication No. WO 98/07880), OPG, and neutrokine-alpha (International Publication No. .
WO
98/1892.1, OX40, and nerve growth factor (NGF), and soluble forms of Fas, CD30, CD27, CD40 and 4-IBB, TR2 (International Publication No. WO 96134.095), DR3 (International Publication No. WO 97/33904), DR4 (International Publication No. WO 98/32856), TRS
(International Publication No: WO 98/30693), TRANK, TR9 (International Publication No.
WO 98/56892),TR10 (International Publication No: WO 98/54202), 312C2 (International w Publication No..WO 98/06842), and TR12, and soluble forms CD154, CD70, and CD153.
' In an additional embodiment, the albumin fusion proteins and/or polynucleotides ~of the invention are administered in combination ~ with angiogenic proteins.
Angiogenic proteins that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Glioma Derived Growth Factor (GDGF), as disclosed in European Patent Number EP-399816; Platelet Derived Growth Factor-A
(PDGF-A), as disclosed in European Patent Number EP-682110; Platelet Derived Growth Factor-B (PDGF-B), as disclosed in European Patent Number EP-282317; Placental Growth Factor (P1GF), as disclosed in International Publication Number WO
92/06194;
Placental Growth Factor-2 (P1GF-2), as disclosed in Hauser et al., Growth Factors, 4:259-268 (1993); Vascular Endothelial Growth Factor (VEGF), as disclosed in International Publication Number WO 90/13649; Vascular Endothelial Growth Factor-A (VEGF-A), as disclosed in European Patent Number EP-50647.7; Vascular Endothelial Growth Factor-2 (VEGF-2), as disclosed in International Publication Number WO 96!39515;
Vascular Endothelial Growth Factor B (VEGF-3); Vascular Endothelial Growth Factor B-186 (VEGF-B186), as disclosed in International Publication Number WO 96/26736;
Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in International Publication Number WO 98/02543; Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in International Publication Number WO 98/07832; and Vascular Endothelial Growth Factor-E
(VEGF-E), as disclosed in German Patent Number DE19639601. The above mentioned references are herein incorporated by reference in their entireties.
In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with Fibroblast Growth Factors.
Fibroblast ' Growth Factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not lirilited to, FGF-1, FGF-2, FGF-3, ' FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, arid FGF-15.
In ~an additional embodiment,..the albumin fusion proteins and/or polynucleotides of the invention ~ are administered in combination with hematopoietic growth factors.
Hematopoietic growth factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited. to, granulocyte macrophage colony stimulating factor (GM-CSF) (sargramostim, LEUKINETM, PROKINET'"), granulocyte colony stimulating factor (G-CSF) (filgrastim, NEUPOGENT"'), macrophage colony stimulating factor (M-CSF, CSF-1) erythropoietin (epoetin alfa, BPOGENTM,~ PROCRITT"'), stem cell, factor (SCF, c-kit ligand, steel factor), megakaryocyte colony stimulating factor, PIXY321 (a GMCSF/IL-3 fusion protein), interleukins, especially any one or more of IL-1 through IL-12, interferon-gamma, or thrombopoietin.
In certain embodiments, albumin fusion proteins and/or polynucleotides of the present invention are administered in combination with adrenergic Mockers, such as, for example, , acebutolol, atenolol, betaxolol, bisoprolol, carteolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, ;propranolol, sotalol, and timolol.
In another embodiment, the albumin fusion proteins and/or 'polynucleotides of the invention are administered in combination with .an antiarrhythmic drug (e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, ,mexiletine,' moricizine, phenytoin, procainamide, N-acetyl procainamide, propafenone, propranolol, guinidine, sotalol, tocainide, and verapamil).
In another embodiment, the albumin fusion proteins andlor polynucleotides of the invention are administered in combination with diuretic agents, such as carbonic anhydrase-inhibiting agents (e.g., acetazolamide, dichlorphenamide, and methazolamide), osmotic diuretics (e.,g., glycerin, isosorbide, mannitol, and urea), diuretics that inhibit Na+-K+-2Cl-symport (e. g., furosemide, bumetanide, azosemide, piretanide, tripamide, ethacrynic acid, muzolimine, and torsemide), thiazide and thiazide-like diuretics (e.g., bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothiazide, . polythiazide, trichormethiazide, chlorthalidone, indapamide, metolazone, and quinethazone), potassium sparing diuretics (e.g., amiloride .and , triamterene), and mineralcorticoid receptor antagonists (e.g., spironolactone, canrenone, ,and potassium canrenoate).
In one embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for endocrine and/or hormone imbalance disorders. Treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, 'z'I, radioactive isotopes of iodine such as 13'I and 'z3I; recombinant growth .hormone, such as HUMATROPET"' (recombinant somatropin); growth hormone analogs such as PROTROPINT"". (somatrem); dopamine agonists such as PARLODEL7"' (bromocriptine); somatostatin analogs such as SANDOSTATINT"" (octreotide);
gonadotropin preparations such as PREGNYLT"', A.P.L.T"" and PROFASIT"" (chorionic gonadotropin (CG)), PERGONALT"" (menotropins), and METRODINT"" (urofollitropin (uFSH));
synthetic human gonadotropin releasing hormone preparations such as FACTRELT"' and LUTREPULSET"" (gonadorelin hydrochloride); synthetic gonadotropin agonists such as LUPRONT"' (leuprolide acetate), SUPPRELINT"" (histrelin acetate), SYNARELT"' (nafarelin acetate), and ZOLADEXT"' (goserelin acetate); synthetic preparations of thyrotropin-releasing hormone such as RELEFACT TRHT"' and THYPINONET"' (protirelin);
recombinant human TSH such as THYROGENT""; synthetic preparations of the sodium salts of the natural isomers of thyroid hormones such as L-TaT"", SYNTHROIDT"' and LEVOTHROIDT"' (levothyroxine sodium), L-T3T"', CYTOMELT"' and TRIOSTATT""
,~
(liothyroine sodium), and THYROLART"" (liotrix); antithyroid compounds such as 6-n-propylthiouracil (propylthiouracil), 1-methyl-2-mercaptoimidazole and TAPAZOLET""
(methimazole), NEO-MERCAZOLET"' (carbimazole); beta-adrenergic receptor antagonists such as propranolol and esmolol; Caz+ channel bloc(eers; dexamethasone and iodinated radiological contrast agents such as TELEPAQUET"' (iopanoic acid) and ORAGRAFINT"' (sodium ipodate).
Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, estrogens or congugated estrogens such as ESTRACET""
(estradiol), ESTINYLT"" (ethinyl estradiol), PREMARINT"', ESTRATABT"', ORTHO-ESTT"', OGENT""
and estropipate (estrone), ESTROVIST"" (quinestrol), EST'RADERMT"' (estradiol), DELESTROGENT"' and VALERGENT"' (estradiol valerate), DEPO-ESTRADIOL
CYPIONATET"" and ESTROJECT LAT"" (estradiol cypionate);. antiestrogens such as NOLVADEXT"' (tamoxifen), SEROPHENET"' and CLOMIDT"' (clomiphene); progestins such as DURALUTINT"' (hydroxyprogesterone caproate), MPAT"' and DEPO-PROVERAT""
(medroxyprogesterone acetate), PROVERAT"' and CYCRINT"' (MPA), MEGACET"' (megestrol acetate), NORLUTINT"' . (norethindrone), and NORLUTATET"' and AYGESTINT"" (norethindrone acetate); progesterone implants such as NORPLANT
SYSTEMT"" (subdermal~ implants of norgestrel); antiprogestins such as RU
486T"' (mifepristone); hormonal contraceptives such as ENOVIDT"" (norethynodrel plus mestranol), PROGESTASERTT"' (intrauterine device that releases progesterone), LOESTRINT"", BREVICONT"', MODICONT"", GENORAT"', NELONAT"', NORINYLT"', OVACON-35T"' and OVACON-50T"" (ethiriyl estradibl/norethindrone), LEVLENT"", NORDETTEr"", TRI-LEVLENT"" and TRIPHASIL-21T"" (ethinyl estradiol/levonorgestrel) LOIOVRALT"' and OVRALT"" (ethinyl estradiol/norgestrel), DEMULENT"" (ethinyl estradiol/ethynodiol diacetate), NORINYLT"", ORTHO-NOVUMT"', NORETHINT"', GENORA'"', and NELOVAT"" (norethindrone/mestranol), DESOGENT"' and ORTHO-CEPTT"" (ethinyl estradiol/desogestrel), ORTHO-CYCLENT"' and ORTHO-TRICYCLENT"' (ethinyl~
2S estradiol/norgestimate), MICRONORT"' and NOR-QDT"' (norethindrone), and OVR>;TTET"' (norgestrel).
Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, testosterone esters such as methenolone .acetate amd testosterone undecanoate; parenteral and' oral androgens such as TESTOJECT-50T""
(testosterone), TESTEXT"' (testosterone propionate), DELATESTRYLT"" (testosterone .
enanthate), DEPO-TESTOSTERONET"" (testosterone cypionate), DANOCRINET"' (danazol), HALOTESTINT"' (fluoxymesterone), ORETON METHYLT"', TESTREDT"' and VIRILONT"' _ 5 (methyltestosterone), and OXANDRINT"" (oxandrolone); testosterone transdermal systems such as TESTODERMT""; androgen receptor antagonist and 5-alpha-reductase inhibitors such as ~ANDROCURT"" (cyproterone acetate), EULEXINT"' (flutamide), and PROSCART"' (finasteride); adrenocorticotropic hormone preparations such as CORTROSYNT""
(cosyntropin); adrenocortical steroids and their synthetic analogs . such as ACLOVATET""
(alclometasone dipropionate), CYCLOCORTT"' (amcinonide), BECLOVENTT"' and VANCERILT"" (beclomethasone dipropionate), CELESTONET"" (betaniethasone), BENISONET"" and UTICORTT"' (betamethasone benzoate), DIPROSONET"' (betamethasone dipropionate), CELESTONE PHOSPHATET"" (betamethasone sodium phosphate), CELESTONE SOLUSPANT"' (betamethasone sodium phosphate and acetate), BETA-VALT"" and VALISONET"' (betamethasone valerate), TEMOVATET"' (clobetasol propionate), CLODERMT"" (clocortolone pivalate), CORTEFT"' and HYDROCORTONET"' , (cortisol (hydrocortisone)); HYDROCORTONE ACETATET"" (cortisol (hydrocortisone) acetate), LOCOIDT"' (cortisoI (hydrocortisone) butyrate), HYDROCORTONE . PHOSPHATET"' (cortisol (hydrocortisone) sodium phosphate), A-HYDROCORTT"" and SOLU
CORTEFT""
(cortisol (hydrocortisone) sodium succinate)-, WESTCORTT"' (cortisol (hydrocortisone) valerate), CORTISONE ACETATET"" (cortisone acetate), DESOWENT"' and TRIDESILONT"" (desonide), TOPICORTT"' (desoximetasone), DECADRONT"' (dexamethasone), DECADRON LAT"' (dexamethasone acetate), DECADRON
PHOSPHATET"' and HEXADROL PHOSPHATET"' (dexamethasone sodium phosphate), FLORONET"' and MAXIFLORT"" (diflorasone diacetate), FLORINEF ACETATET""
(fludrocortisone acetate), AEROBIDT"" and' NASALIDET"' (flunisolide), FLUONIDT"' and 172 ' .' SYNALART"" (fluocinolone acetonide), LIDEXT'" (fluocinonide), FLUOR-OPT"' and FMLT"' (fluorometholone), CORDRANT"" (flurandrenolide), _HALOGT"' (halcinonide), HMS
LIZUIFILMT"' (medrysone), MEDROLT"' (methylprednisolone), DEPO-MEDROLT"' and MEDROL ACETATET"' (methylprednisone acetate), A-METHAPREDT"' and SOLUMEDROLT"' (methylprednisolone sodium succinate), ELOCONT"' (mometasone furoate), HALDRONET"' (paramethasone acetate), DELTA-CORTEFT"' (prednisolone), ECONOPREDT"" (prednisolone acetate), HYDELTRASOLT"" (prednisolone sodium phosphate), HYDELTRA-T.B.AT"" (prednisolone tebutate), DELTASONET'"
(prednisone), ARISTOCORTT"" and KENACORTT"' (triamcinolone), KENALOGT"' (triamcinolone IO acetonide), ARISTOCORTT"' and KENACORT DIACETATE'"' (triamcinolone diacetate), and ARISTOSPANT"' (triamcinolone hexacetonide); inhibitors of biosynthesis and action of adrenocortical steroids such as CYTADRENT"' (aminoglutethimide), NIZORALT""
(ketoconazole), MODRASTANET"' ~ (trilostane), and METOPIRONET"' (metyrapone);
bovine, porcine or human insulin or mixtures thereof; insulin analogs;
recombinant human 1S insulin such as HUMULINT"' and NOVOLINT""; oral hypoglycemic agents such as ORAMIDET"' and ORINASE~"' (tolbutamide), DIABINESET"' (chlorpropamide), TOLAMIDET"' and TOLINASET"" (tolazamide), DYMELORT"' (acetohexamide), glibenclamide, ' MICRONASET"', DIBJJTAT"' and GLYNASET"' (glyburide), , GLUCOTROLT"' (glipizide), and DIAMICRONT"' (gliclazide), GL,UCOPHAGET""
20 (metformin), ciglitazone, pioglitazone, and alpha-glucosidase inhibitors;
bovine or porcine glucagon; somatostatins such as SANDOSTATINT"' (octreotide);~ and diazoxides such as PROGLYCEMT"' (diazoxide).
In one embodiment, the albumin fusion proteins andlor polynucleotides of the invention are administered in combination with treatments for uterine motility disorders.
25 Treatments for uterine motility disorders include,.but are not limited to, estrogen drugs such as conjugated estroger~s (e.g., PREMARIN~ and ESTRATAB~), estradiols' (e.g., CLIMARA~ and ALORA~), estropipate, and chlorotrianisene; progestin. drugs (e.g., WO 01/79442 ~ PCT/USO1/11850 AMEN° (medroxyprogesterone), 1VIICRONOR° (norethidrone acetate), PROMETRIUM°
progesterone, and megestrol acetate); and estrogen/progesterone combination therapies such as, for example, conjugated estrogens/medroxyprogesterone (e.g., PREMPROT""
and PREMPHASE°) and norethindrone acetate/ethinyl estsradiol (e.g., FEMHRTT"').
In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in.combination with drugs effective in treating iron deficiency and hypochromic anemias, including but not limited to, ferrous sulfate (iron sulfate, FEOSOLTM), ferrous fumarate (e.g., FEOSTATTM), ferrous gluconate (e.g., FERGONT"'), polysaccharide-iron complex (e.g., NIFEREXTM), iron dextran injection (e.g.;
INFEDTm), cupric sulfate, , pyroxidine, riboflavin, Vitamin Blz, cyancobalamin injection (e.g., REDISOLT"', RUBRAMIN PCT'"), hydroxocobalamin, folic acid (e.g., FOLVITETM), leucovorin (folinic acid, 5-CHOH4PteGlu, citrovorum factor) or WELLCOVORIN
(Calcium salt of leucovorin), transferrin or feri:itin.
In certain embodiments, the albumin fusion proteins and/or polynucleotides lof the invention are administered in combination with agents used. to treat psychiatric disorders.
Psychiatric drugs that may be administered with the albumin fusion proteins and/or .
polynucleotides of the invention include, but are not limited to, antipsychotic ageilts (e.g., chlorpromazine, chlorprothixene, clozapine, fluphenazine, haloperidol, loxapine, mesoridazine, molindone, olanzapine, perphenazine, pimozide, quetiapine, risperidorie, thioridazine, thiothixene, trifluoperazine, and triflupromazine), antimanic agents (e.g., carbamazepine, divalproex sodium, lithium carbonate, and lithium citrate), antidepressants (e.g., amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin, fluvoxamine, fluoxetine, imipramine, isocarboxazid, maprotiline, mirtazapine, nefazodone, nortriptyline, paroxetine, phenelzine, protriptyline, sertraline, tranylcypromine, trazodone, trimipramine, and venlafaxine), antianxiety agents (e.g., alprazoIam, buspirone, chlordiazepoxide, clorazepate, diazepam, halazepam, lorazepam, oxazepam, and prazepam), and stimulants (e.g., d=amphetamine, methylphenidate, and pemoline).
In other embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat neurological disorders.
Neurological agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antiepileptic agents (e.g., - carbamazepine, clonazepam, ethosuximide, phenobarbital, phenytoin, primidone; valproic acid, divalproex sodium, felbamate; gabapentin, lamotrigine, levetiracetam, oxcarbazepine, tiagabine, topiramate, zonisarilide, diazepam, lorazepam, and clonazepam), ~antiparkinsonian agents (e.g., .levodopa/carbidopa, selegiline, amantidine, bromocriptine, pergolide, , ropinirole, pramipexole, benztropine; biperiden; ethopropazine; procyclidine;

trihexyphenidyl, tolcapone), and ALS therapeutics (e.g. riluzole).
In another embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with vasodilating agents and/or calcium chailnel blocking agents. Vasodilating agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include,- but are not limited to, Angiotensin Converting Enzyme (ACE) inhibitors (e.g., papaverine, isoxsuprine, benazepril, captopril, cilazapril, enalapril, enalaprilat, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, spirapril, trandolapril, and nylidrin), and nitrates (e:g., isosorbide dinitrate, isosorbide mononitrate, and nitroglycerin). Examples of calcium channel blocking agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to amlodipine, bepridil, diltiazem, felodipine, flunarizine, isradipine, nicardipine, nifedipine, nimodipine, and verapamil. - ~ .
In certain embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered. in combination with treatments for gastrointestinal disorders.
Treatments for gastrbintestinal disoi~ders that may be administered with the albumin fusion protein and/or polynucleotide of the invention include, but are not limited to, HZ histamine receptor antagonists (e.g., TAGAMETT"' (cimetidine), ZANTACTM (ranitidine), PEPCIDTM
(famotidine), and AXIDTM (nizatidine)); inhibitors of H+, K+ ATPase (e.g., PREVACIDTT'' (lansoprazole) and PRILOSECTM (omeprazole)); Bismuth compounds (e.g., PEPTO-BISMOLT"' (bismuth subsalicylate) and DE-NOLTM (bismuth subcitrate)); various antacids;
sucralfate; prostaglandin~analogs (e.g. CYTOTECT"'' (misoprostol)); muscarinic cholinergic antagonists; laxatives (e.g.; surfactant laxatives, stimulant laxatives, saline and osmotic laxatives); antidiarrheal agents (e.g., LOMOTILT°'' (diphenoxylate), MOTOFEN~'~' (diphenoxin), and IMODIUMTM (loperamide hydrochloride)), synthetic analogs of somatostatin such as SANDOSTATINTM (octreotide), antiemetic agents (e.g., ZOFRAN~'T'' (ondansetron), KYTRILTM (granisetron hydrochloride), tropisetron, ~
dolasetron, metoclopramide, chlorpromazine, perphenazine, prochlorperazine, promethazine, . thiethylperazine, triflupromazine, domperidone, haloperidol, droperidol, trimethobenzamide, dexamethasone, methylprednisolone, dronabinol~ and nabilone); DZ
antagonists (e.g., metoclopramide, trimethobenzamide and chlorpromazine); bile salts;
chenodeoxycholic acid; ursodeoxycholic acid; and pancreatic enzyme preparations such as pancreatin and pancrelipase. .
In additional embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with other ' therapeutic or prophylactic regimens, such as, for example, radiation therapy.

The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions comprising . albumin fusion proteins of the invention. Optionally associated with such containers) can be a notice in the form prescribed.by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
Gene Therany , Constructs encoding albumin fusion proteins of the invention can be used as a part of a gene therapy protocol to deliver therapeutically effective doses of the albumin fusion protein. A preferred approach for i~ vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, encoding an albumin fusion protein of the invention.
Infection of cells with a~viral vector has the advantage that a large proportion of the targeted -15 . cells can receive the nucleic acid. Additionally, molecules encoded within the viral 'vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid.
Retrovirus vectors and adeno-associated virus vectors can be used as a recombinant gene delivery system for the transfer of exogenous nucleic acid molecules encoding albumin fusion proteins in vivo: These vectors provide efficient delivery of nucleic acids into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host. The development of specialized cell lines (termed "packaging cells") which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A.D. (1990) Blood 76:27 1). A replication defective retrovirus can be packaged into virions which can be used to infect a target cell through the use ,of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current,Protocols in Molecular Biology, Ausubel, F.M. et al., (eds.) Greene Publishing , Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.
Another viral gene delivery system useful in the present invention uses adenovirus-derived vectors. The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate.in a normal lytic viral life cycle. See, for example, Berkner et al., 'BioTechniques 6:616 (1988); Rosenfeld et al:, Science 252:431-434 (1991); and Rosenfeld et al., Cell 68:143-155 (1992). Suitable adenoviral vectors derived~from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are known to those 176 .

skilled in the art. Recombinant adenoviruses can be advantageous in certain circumstances in that they are not capable of~infecting nondividing cells and can be used to infect a wide variety of cell types, including epithelial cells (Rosenfeld et al., (1992) cited supra).
Furthermore,- the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity. .
Additionally, introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into, the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g.= retroviral -DNA). Moreover, the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al., cited supra; Haj-Ahmand et al., J.
Virol. 57:267 (1986)).
.~ .In another embodiment, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject nucleotide molecule by the targeted 1S cell. Exemplary gene delivery systems of this type include liposomal derived systems;
poly-lysine conjugates, and artificial viral envelopes. In a representative embodiment, a nucleic acid molecule encoding.an albumin fusion protein of the invention can be entrapped in liposomes bearing positive charges on their surface (e.g.= lipofectins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et al. (1992) No Shinkei Geka 20:547-5 5 l; PCT publication W091/06309; Japanese patent application 1047381;~and European patent publication EP-A-43075).
Gene delivery systems for a gene encoding an albumin fusion protein of the invention can be introduced into a patient by any of a number of methods. For instance, a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g.
by intravenous injection, and specific transduction of the protein in. the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof. In other embodiments, initial delivery of the re=combinant gene is more limited with introduction into the animal being quite localized. For example,, the gene delAvery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by Stereotactic injection (e.g. Chen et al. (1994) PNAS 91: 3 054-3 OS 7). The pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable. diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Where the albumin fusion protein can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the' albumin fusion protein. .
. ' 177 WO 01/79442 ~ PCT/USO1/11850 Additional Gene Therapy Methods Also encompassed by the invention are gene therapy methods for treating or preventing disorders, diseases and conditions. The gene therapy methods relate to the introduction of nucleic acid (DNA, RNA and antisense DNA or RNA) sequences into an animal to achieve expression of an albumin fusion protein of the invention.
This method requires a polynucleotide which codes for an albumin fusion protein of the present invention operatively linked to a promoter and any other genetic elements necessary 'for the expression of the fusion protein by the target tissue. Such gene therapy and delivery techniques are known . in the art, see, for example, W090/11092, which is herein incorporated by reference.
Thus, for example, cells from a patient may be engineered with a polynucleotide (DNA or RNA) comprising a promoter operably linked to a polynucleotide encoding an albumin fusion protein of the present invention ex vivo, with the engineered cells than being provided to a patient to be treated with the fusion protein of the present invention.
Such methods are well-known in the art. For example, see Belldegrun, A., et al., J. Natl.
Cancer Inst. 85: 207-216 (1993); Ferrantini, M. et al., Cancer Research 53:
1_107-1112 (1993); Ferrantini, M. et al., J. Immunology 153: 4604-4615 (1994); I~aido, T., et al., Int..
J. Cancer 60: 221-229 (1995); Ogura, H., et al., Cancer Research 50: 5102-5106 (1990);
Santodonato, L., et al., Human Gene Therapy 7:1-10 (1996); Santodonato, L., et al., Gene Therapy 4:1246-1255 (1997); and Zhang, J.-F. et al., Cancer Gene Therapy 3: 31-(1996)), which are herein incorporated by reference. In one embodiment, the cells which . are engineered are arterial cells. The arterial cells may be reintroduced into the patient through direct injection to the artery, the tissues surrounding the artery, or through catheter injection.
As discussed in more detail below, the polynucleotide constmcts can be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, and the like). The polynucleotide constructs may be delivered in a pharmaceutically acceptable liquid or aqueous carrier.
In one embodiment, polynucleotides encoding the albumin fusion proteins of the present invention is delivered as a naked polynucleotide. The term "naked"
polynucleotide, , DNA or~RNA refers to sequences that are free from any delivery vehicle that acts to assist, .
promote or facilitate entry into the cell, including viral sequences,, viral particles, liposome formulations, lipofectin or precipitating agents and the like. However, polynucleotides encoding the albumin fusion proteins of the present invention can also be delivered in liposome formulations and lipofectin formulations and the like can be prepared by methods well known to those skilled in the art. Such methods are described, for example, in U.S.
Patent Nos. 5,593,972, 5,589,466, and 5,580,859, which are herein incorporated by reference.
The polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication. Appropriate vectors include pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; pSVK3, pBPV, pMSG and pSVL available from ,.
Pharmacia; and pEFI/VS,.pcDNA3.1, and pRc/CMV2 available from Invitrogen.
Other suitable vectors will be readily apparent to the skilled artisan.' ~ .
Any strong promoter known to those skilled in the art can be used for driving the expression of the polynucleotide sequence. Suitable promoters include adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter;
inducible promoters, such as the MMT~ promoter, the metallothionein promoter;
heat shock IS promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter;
retroviral LTRs; the b-actin promoter; and human growth hormone promoters. The promoter also may be the native promotes for the gene corresponding to the' Therapeutic protein portion of the albumin fusion proteins of the invention.
Unlike other gene .therapy techniques, one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide .
synthesis in the cells. Studies have shown that non-replicating DNA sequences 'can be introduced into cells to provide production of the desired polypeptide for periods of up to six months.
The polynucleotide construct can be delivered to the interstitial space of tissues within the an animal, including of muscle, skin, brain, lung, liver, spleen;
bone marrow, v thymus, heart, Iyrriph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, .and connective tissue.
Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within.connective tissue ensheathing' muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels. Delivery to~the interstitial space of muscle tissue is preferred for the, reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, WO 01/79442 . PCT/USO1/11850 such as, for example, stem cells of blood or skin fibroblasts. l~ vivo muscle cells axe particularly competent in their ability to take up and express polynucleotides.
For the naked nucleic acid sequence injection, an effective dosage amount of DNA
or RNA will be 'in the range of from about 0.05 mg/kg body weight to abciut 50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg=to about S mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can.readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration.
The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. However, ~ other parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for,delivery to lungs or bronchial tissues, throat or mucous membranes of the nose. In addition; naked DNA constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.
The naked polynucleotides are delivered by any method known in the art, including, but not limited to, direct needle injection .at the delivery site, intravenous injection, topical administration, catheter infusion, and so-called "gene guns". These delivery methods are known in the art.
The constructs may also be delivered with delivery vehicles such as viral sequences, viral particles, liposome formulations, lipofectin, precipitating agents, etc.
Such methods of delivery are known in the art.
In certain embodiments, the polynucleotide constructs are complexed in a liposome preparation. Liposomal preparations for use ,.in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations.
However, cationic li,posomes are particularly preferred because a tight charge complex can be formed between the cationic liposome and the polyanionic ilucleic acid. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al., Proc. Natl. ' Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference); mRNA
(Malone et. al., Proc. Natl. Acad. Sci. USA (1989) 86:6077-6081, which is herein incorporated by, reference); and purified transcription factors (Debs et al., J. Biol. Chem.
(1990) 265:10189-10192, which is herein incorporated by reference), in functional form.
Cationic ~ liposomes , are - readily available. , For example, N[1-2,3-dioleyloxy)propyl~-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N:Y. (See, also, ~Felgner et al., Proc. Natl Acad. Sci. USA
(1987) 84:7413-7416, which is herein incorporated by reference). Other commercially available liposomes include transfectace (DDABIDOPE) and DOTAP/DOPE (Boehringer).
Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication No. WO 90/11092 (which is herein incorporated by reference) for a description of the synthesis of DOTAP
, ( 1,2-bis(oleoyloxy)-3-(trimethylammonio)propane) Iiposomes. Preparation of DOTMA
liposomes is explained in the literature, see, e.g., P. Felgner et al., Proc.
Natl. Acad. Sci.
USA 84:7413-7417, which is herein incorporated by reference. Similar methods can be used to prepare liposomes from other cationic lipid materials.
Similarly, anionic and neutral liposocmes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials.
Such materials include phosphatidyl, choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol . (DOPG), dioleoylphoshatidyl ethanolamine . (DOPE), among others. These materials can also be mixed with the DOTMA and DOTAP starting materials .in appropriate .ratios.
Methods for making 1'iposomes using these materials are well known in the art.
For example, commercially dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine (DOPE) can be used in various combinations to make conventional liposomes, with or ~
without the addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can be prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water. The sample is then sonicated for 2 hours in .a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15EC. Alternatively, negatively charged ~ -vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion thrbugh nucleopore membranes to produce unilamellar vesicles of discrete size.
Other , methods are known and available to those of skill in the art.
The liposomes can corrzprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred. The various liposome-nucleic acid complexes are prepared using methods well known in the art.
See, e.g., Straubinger et al., .Methods of Immunology (1983), 101:512-527, which is herein incorporated by reference. For example, MLVs containing nucleic acid can be prepared by depositing. a thin film of phospholipid on the walls of a glass tube and subsequently hydrating with a solution of the material to. be encapsulated.
SUVs are prepared by extended sonication of MLVs to p>:oduce a homogeneous population of unilamellar liposomes. The material to be entrapped is added to a suspension of preformed MLVs and then sonicated. When using liposomes containing cationic lipids, the dried lipid .

film is resuspended in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCI, sonicated, and then the preformed liposomes are mixed directly with the DNA. The liposome and DNA fornn a very stable complex due to binding of the positively charged liposomes to the cationic DNA. SUVs find use with small nucleic acid fragments. LUVs are prepared by a number of methods, well known in the art. , Commonly used methods include Ca2+-EDTA chelation (Papahadjopoulos et al., Biochim.
Biophys. Acta (1975) 394:483; Wilson et al., Cell 17:77 (1979)); ether injection (Deamer, D. and Bangham, A., Biochim. Biophys. Acta 443:629 (1976); Ostro et al., Biochem.
Biophys. Res. Commun. 76:836 (1977); Fraley et al.,~Proc. Natl. Acad. Sci. USA
76:3348 (1979)); detergent dialysis (Enoch, H. and Strittmatter, P., Proc. Natl. Acad.
Sci,. USA.
76:145 (1979)); and reverse-phase evaporation (REV) (Fraley et ~al., J. Biol.
Chem.
255:10431 (1980); Szoka, F. and Papahadjopoulos, D., Proc. Natl. Acad. Sci.
USA
75:145 (1978); Schaefer-Ridder et. al., Science 215:166 (1982)), which are herein incorporated by reference.
Generally, the ratio of DNA to 1'iposomes will be from about 10:1 to about 1:10.
Preferably, the ration will be from about 5:1 to about 1:S. More preferably,.
the ration will be about 3:1 to about I:3. Still more preferably, the ratio will be about 1:
I.
U.S. Patent No. 5,676,954 (which is herein incorporated by reference) reports on the injection of genetic material, complexed with cationic liposomes carriers, into mice.
U.S. Patent Nos. 4,897,355, 4,946,787, 5,049,386, 5,459,127, 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 (which are herein incorporated by reference) provide cationic lipids for use in transfecting DNA
into cells and mammals. U.5. Patent Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 provide methods for delivering DNA-cationic lipid complexes to mammals.
Tn certain embodiments, cells are engineered, ex vivo or i~ vivo, using a retroviral , particle containing RNA which comprises a sequence encoding an albumin fusion protein of the present invention. Retroviruses from which the retroviral plasmid vectors may be _ derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Rous sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected-include, but are not limited to, the PE501, PA317, R-2, R-AM, PA12, T19-14X, VT-19-17-H2, RCRE, RCRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, Human Gene . Therapy 1:5-14 (1990), which is incorporated herein by reference in its entirety. The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaP04 precipitation. In one alternative, the retroviral plasmid vector may-be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
The producer cell line generates infectious retroviral vector particles which include polynucleotide encoding an albumin fusion protein of the present invention.
Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express a fusion protin of the present invention.
In certain other embodiments, cells are . engineered, ex vivo or in vivo, with polynucleotide contained in an adenovirus vector. Adenovirus can be manipulated such that it encodes and expresses fusion protein of the present invention, and at the same time is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. Adenovirus expression is achieved without integration of the viral DNA into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis. Furthermore, adenoviruses have been used as live enteric vaccines for many .years with an excellent safety profile (Schwartz et al. Am. Rev. Respir: Dis.109:233-238 (1974)). Finally, adenovirus mediated gene transfer has been demonstrated in a number of instances including transfer of alpha-1-antitrypsin and CFTR to the lungs of cotton. rats (Ros~nfeld, M. A. et a1. (1991) .
Science~252:431-434; Rosenfeld et al., (1992) Cell 68:143-155). Furthermore, extensive studies to attempt to establish adenovirus as a causative agent in human cancer were uniformly negative (Green, M. et al. (1979) Proc.~Natl. Acad. Sci. USA
76:6606).
Suitable adenoviral vectors useful in the present invention are described, for example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel. 3:499-503 (1993);
Rosenfeld et al., Cell 68:143-155 (1992); Engelhardt et al., Human Genet. Ther. 4:759-769 (1993);
Yang et al.,. Nature Genet. 7:362-369 (1994); Wilson et al., Nature 365:691-692 (1993);
and U.S. Patent No. 5,652,224, which are herein incorporated by reference. For example, the adenovirus vector Ad2 is useful and can be grown in human 293 cells. These cells contain , the E1 region of adenovirus and constitutively express Ela and Elb, which complement the defective adenoviruses by providing the products of the genes deleted from the vector. In addition~to Ad2, other varieties of adenovirus (e.g., Ad3,~
AdS, and Ad7) are also useful in the present invention.
Preferably, the adenoviruses used in the present invention are replication deficient.
Replication deficient adenoviruses require the aid of a helper virus andlor packaging cell line to form infectious particles. The resulting virus is capable of infecting cells and can express , a polynucleotide of interest which is operably linked to a promoter, but cannot replicate in most cells. Replication deficient adenoviruses may be deleted in one or more of all or a portion of the following genes: Ela, Elb, E3, E4, E2a, or Ll through L5.

In certain other embodiments, the cells are engineered, ex vivo or in viv~, using an adeno-associated virus (AAV). AAVs are naturally occurring defective viruses that require helper viruses to produce infectious particles (Muzyczka, N.; Curr. Topics in Microbiol.
Immunol. 158:97 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells. Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate, but space for exogenous DNA is limited to about 4.5 kb.
Methods for producing and using such AAVs are known in the art. See,. for example, U.S.
Patent Nos.
5,139,941,.5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and 5,589,377.
For example, an appropriate AAV vector for use in the present invention will include all the sequences necessary for DNA replication, encapsidation, and host=cell integration.
The polynucleotide construct is inserted into the AAV vector using standard cloning methods, such as those found in Sambrook et al., Molecular Cloning: A
Laboratory Manual, Cold Spring Harbor Press (1989). The recombinant AAV vector is then transfected into packaging cells which are infected with a helper virus, using any standard technique, including lipofection, electroporation, calcium phosphate precipitation, etc.
Appropriate helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or herpes viruses. ~ Once the packaging cells are transfected and infected, they will produce infectious AAV viral particles which contain the polynucleotide .construct.
These viral particles are then used to transduce eukaryotic cells, either ex vivo or i~c vivo. The transduced cells will contain the polynucleotide construct integrated into its . genome, and will express a fsuion protein of the invention.
Another method of gene therapy involves operably associating heterologous control , regions and endogenous polynucleotide sequences (e.g. encoding a polypeptide of the present invention) via homologous recombination (see, e.g., U.S. Patent No.
5,641,670, issued June 24, 1997; International Publication No. WO 96/29411, published September 26, 1996; International Publication No. WO 94/12650, published August 4, 1994;
Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), which are herein encorporated by reference. This method involves the activation of a gene which is present in the target cells, but which is not normally expressed in the cells, or is expressed at a lower level than desired.
Polynucleotide constructs are made, using standard techniques known in the art, which contain the promoter with targeting sequences flanking the promoter.'-Suitable promoters are described herein. The targeting sequence is sufficiently complementary to an endogenous sequence to permit homologous recombination of the promoter-targeting sequence with the endogenous sequence. The~fargeting sequence will be sufficiently near the 5' end of the desired endogenous polynucleotide sequence so the -promoter will be operably linked to the endogenous sequence upon homologous recombination.

The promoter and the targeting sequences can be amplified using PCR.
Preferably, the amplified promoter contains distinct restriction enzyme sites on the 5' and 3' ends.
Preferably, the 3' end of the first targeting sequence contains the same restriction enzyme site as the 5' end of the amplified promoter and the 5' end of the second targeting sequence.
contains the same restriction site as the 3' end of the amplified promoter.
The amplified promoter and targeting sequeilces are digested and.ligated together. ' The promoter-targeting sequence construct is delivered,to the cells, either as naked polynucleotide, or in conjunction with transfection-facilitating agents, such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc., described in more detail~above. The P promoter-targeting sequence can be delivered by any method, included direct needle injection, intravenous injection, topical administration, ' catheter infusion, particle accelerators, etc. The methods are described in more detail below.
The promoter-targeting sequence construct is taken up by cells. Homologous recombination between the construct and the endogenous sequence takes place, such that an' 15. endogenous sequence is placed under the control of the promoter. The promoter then drives the expression of the endogenous sequence.
The polynucleotide encoding an albumin fusion protein of the present invention may /
contain a secretory signal sequence that facilitates secretion of the protein.
Typically, the .
signal sequence is positioned in the coding region of the polynucleotide to be expressed towards or at the 5' end of the coding region. The signal sequence may be homologous or heterologous to the polynucleotide of interest and may be homologous or heterologous to the cells to be transfected. Additionally, the signal sequence may be chemically synthesized using methods known in the art.
Any mode of administration of any of the above-described polynucleotides constructs can be used so long as the mode results in the expression of one or more molecules in an amount sufficient to provide a therapeutic effect.. This includes direct needle injection, systemic injection, catheter infusion, biolistic injectors, particle accelerators (i.e., "gene 'guns"), gelfoam sponge depots, other commercially available depot materials, ' osmotic pumps (e.g., Alza minipumps), .oral or suppositorial solid (tablet or pill) 30. pharmaceutical formulations, and, decanting or topical applications during surgery. For example, direct injection of naked calcium phosphate-precipitated plasmid into rat liver and rat spleen or a protein-coated plasmid into the portal vein has resulted in gene expression of - the foreign gene in the rat livers (Kaneda et al., Science 243:375 (1989)).
A preferred method of local administration is by direct injection. Preferably, ~an albumin fusion protein of the present invention complexed with a delivery vehicle is .
administered by direct injection into or locally within the area of arteries.
Administration of a composition locally within the area of arteries refers to injecting the.
composition centimeters ' 185 , and preferably, millimeters within arteries.
Another method of local adnninistration is to contact a polynucleotide construct of the present invention in or around a surgical wound. For example, a patient . can undergo surgery and the polynucleotide construct can be coated on the surface of tissue inside the wound or the construct can be injected into areas of tissue inside the wound.
Therapeutic compositions useful in systemic administration, include fusion proteins of the present invention complexed to a targeted delivery vehicle of the present invention.
Suitable delivery vehicles for use with systemic administration comprise liposomes comprising ligands for targeting the vehicle to a particular site. In specific embodiments, suitable delivery vehicles for use with systemic administration comprise liposomes comprising albumin fusion proteins of the invention for targeting the vehicle to a particular site.
Preferred. methods of systemic administration, include intravenous injection, aerosol, oral and percutaneous (topical) delivery. Intravenous injections can be performed - 15 using methods standard in the art. Aerosol delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad.
Sci. USA
189:11277-11281, 1992, which is incorporated herein by reference). Oral delivery can be performed by complexing a polynucleotide construct of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art. Topical delivery can be performed by mixing a polynucleotide construct of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin.
Determining an effective amount of substance to be delivered can depend upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the animal, the precise condition requiring treatment and its severity, and the route of administration. The frequency of treatments depends upon a number of factors, such as the amount of polynucleotide constructs administered per dose, as well as the health and history of the subject. The precise amount, number of doses, and timing of doses~will be determined by the attending physician or.
veterinarian.
~ Albumin fusion proteins of the present invention can be administered to any animal, preferably to mammals and birds. Preferred mammals include humans, dogs, cats, mice, rats, rabbits sheep, cattle, horses and pigs, with humans being particularly preferred.
Biological Activities Albumin fusion proteins and/or polynucleotides encoding albumin fusion proteins of the present invention, can be used in assays to test for one or more biological activities. If WO 01/79442 _ PCT/USO1/11850 -an albumin fusion protein and/or polynucleotide exhibits an activity in a particular assay, ,it is likely that the Therapeutic protein corresponding to the fusion portein may be involved in the diseases associated with the biological activity. Thus the fusion protein could be used to treat the associated disease. . _ Members of the secreted family of proteins are believed to be involved in biological activities associated with, for example, cellular signaling. Accordingly, albumin fusion proteins of the invention and polynucleotides encoding these protiens, may be used in diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders associated with aberrant activity of secreted polypeptides.
In preferred embodiments, fusion proteins of the present invention may be used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders relating to diseases and disorders of the endocrine system, the nervous system (See, for example, "Neurological Disorders" section below), the immune system (See, for example, "Immune Activity" section below), respiratory system (See, for example, "Respiratory Disorders"
section below), cardiovascular system (See, for example, "Cardiovascular Disorders"
section below), reproductive system (See, for example, "Reproductive System Disorders"
section below) digestive system (See, for example, "Gastrointestinal Disorders" section below), diseases aridlor disorders relating to cell proliferation (See, for example, "Hyperproliferative Disorders/Cancer" section below), and/or diseases or disorders relating to the blood ((See, for example, "Blood-Related Disorders" section below).
In preferred embodiments, the present invention encompasses a method of treating a y 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 portion corresponding to a Therapeutic protein 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.
In certain embodiments, an albumin fusion protein of the present invention may be used to diagnose and/or prognose diseases and/or disorders associated with the tissues) in which the gene corresponding to the Therapeutic protein portion of the fusion portien of 'the invention is expressed.
Thus, fusion proteins of the invention and polynucleotides~encoding albumin fusion proteins of the invention are useful in the diagnosis; detection and/or treatment of diseases and/or disorders associated with activities that include, but are not limited to, prohormone .
activation, neurotransmitter activity, cellular signaling, cellular .proliferation, cellular differentiation, and cell migration.
187 .

More generally, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful for the diagnosis, prognosis, prevention andlor treatment of diseases and/or disorders associated with the following systems.
Immune Activity Albumin fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing diseases, disorders, and/or conditions of the immune system,~by, for example, activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of immune cells. Immune cells develop.through a process called hematopoiesis, producing myeloid (platelets, red~blood cells, neutrophils, and macrophages) and lymphoid (B and T ' lymphocytes) cells from plu ripotent stem cells. The etiology of these immune diseases, disorders, and/or conditions maybe genetic, somatic, such as cancer and some autoimmune .
diseases, acquired (e.g., by 'chemotherapy or toxins), or infectious.
Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as a marker or detector of a particular immune system disease or disorder In another embodiment, a' fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention, may be used to treat diseases and .
disorders of the immune system andlor to inhibit or enhance an immune response generated by cells associated with the tissues) in which the polypeptide of the invention is expressed.
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing, and/or prognosing immunodeficiencies, including ' both congenital and acquired .
immunodeficiencies. Examples of B cell immunodeficiencies in which immunoglobulin levels B cell function and/or B cell numbers , axe decreased' include: X-linked agammaglobulinemia (Bruton's disease), X-linked infantile agammaglobulinemia, X-linked immunodeficiency, with hyper IgM, non X-linked immunodeficiency with hyper IgM, X-linked lymphoproliferative .syndrome (XLP), agammaglobulinemia including congenital and acquired agammaglobulinemia, adult onset agammaglobulinemia, late-onset agammaglobulinemia, dysgammaglobulinemia, hypogammaglobulinemia, unspecified hypogammaglobulinemia, recessive agammaglobulinemia (Swiss type), Selective IgM
deficiency, selective IgA 'deficiency, selective IgG subclass deficiencies, IgG subclass deficiency (with or without IgA deficiency), Ig deficiency with increased IgM, IgG 'and IgA
deficiency with increased IgM; antibody deficiency with normal or elevated Igs, Ig heavy chain deletions, kappa chain , deficiency, B cell lymphoproliferative disorder (BLPD), WO 01/79442 . PCT/USO1/11850 common variable immunodeficiency (CVID), common variable immunodeficiency (CVI) (acquired), and transient hypogammaglobulinemia of infancy.
In specific embodiments, ataxia-telangiectasia or conditions associated with ataxia telangiectasia are treated, prevented, 'diagnosed, and/or prognosing using the, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
EXamples of congenital immunodeficiencies in which T cell and/or B cell function and/or number is decreased include, but are not limited to: DiGeorge anomaly, severe combined immunodeficiencies (SLID) (including, but not limited to, X-linked SCID, IO autosomal recessive SCID, adenosine deaminase deficiency, purine nucleoside ~phosphorylase (PNP) deficiency, Class II MHC deficiency (Bare lymphocyte syndrome), Wiskott-Aldrich ~syndrorne, and ataxia telangiectasia), thymic hypoplasia, third and fourth pharyngeal pouch syndrome, 22q 11.2 deletion, chronic mucocutaneous candidiasis, natural killer cell deficiency (NK), idiopathic CD4+ T-lyniphocytopenia, .immunodeficiency with predominant T cell defect (unspecified), and unspecified immunodeficiency of cell mediated immunity.
In specific embodiments, DiGeorge anomaly or conditions associated with DiGeorge anomaly are treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
Other immunodeficiencies that may be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, chronic grariulomatous disease, Chediak-Higashi ,syndrome, myeloperoxidase deficiency, leukocyte glucose-6-phosphate dehydrogenase deficiency, X-linked lymphoproliferative syndrome (XLP), leukocyte adhesion deficiency; complement component deficiencies (including C1, C2, C3, C4, C5, C6, C7, C8 and/or C9 deficiencies), reticular dysgenesis, thymic alymphoplasia aplasia, immunodeficiency with thymoma, severe congenital leukopenia, dysplasia with immunodeficiency, neonatal neutropenia, short limbed dwarfism, and Nezelof syndrome . combined immunodeficiency with Igs.
In a preferred embodiment, the immunodeficiencies and/or conditions associated with the iinmunodeficiencies recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. , ' In a preferred embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among immunodeficient individuals. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among B
cell and/or T cell immunodeficient individuals.
The albumin fusion proteins. of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing ,autoimmune disorders. Many autoimmune disorders result from inappropriate recognition of self as foreign material by immune cells. This inappropriate recognition results in an immune response leading to the destruction of the host tissue.
Therefore, the administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention ~ that can inhibit an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing autoimmune disorders.
Autoimmune diseases or disorders that may be treated, prevented, diagnosed and/or prognosed by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, one or more of the following:
systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, autoimmune thyroiditis, Hashimoto's thyroiditis, autoimmune hemolytic anemia, w hemolytic anemia, thrombocytopenia, autoimmune thrombocytopenia purpura, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, purpura (e.g., Henloch-Scoenlein purpura), autoimmunocytopenia, Goodpasture's syndrome, Pemphigus vulgaris, myasthenia' gravis, Grave's disease (hyperthyroidism), and insulin-resistant diabetes mellitus.
Additional disorders that are likely to have an autoimmune component that may be treated, prevented, and/or diagnosed with the albumin fusion proteins of the , invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are . not limited to, type II collagen-induced arthritis, antiphospholipid syndrome, dermhtitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, ~ rheumatic heart disease, neuritis, uveitis ophthalmia, ~polyendocrinopathies, Reiter'-.s Disease, Stiff-Man Syndrome, autoimmune pulmonary inflammation, autism, Guillain-Barre Syndrome, insulin dependent diabetes mellitus, and autoimmune inflammatory eye disorders.
Additional disorders that are likely to have an autoimrimne component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease .
(often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis (often characterized, e.g., by nonhistone ANA), pernicious anemia (often-characterized, e.g., by antiparietal cell, microsomes, and intrinsic factor antibodies), WO 01/79442 ' PCT/USO1/11850 idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized, e.g., by antispermatozoal antibodies), glomerulonephritis (often characterized, e.g., by glomerular basement membrane antibodies or immune complexes), bullous pemphigoid (often characterized, e.g., by IgG
and complement in basement membrane),. Sjogren's syndrome (often characterized, e.g., by multiple tissue antibodies, and/or a specific nonhistone ANA (SS-B)), diabetes mellitus (often characterized, e.g., by cell-mediated and humoral islet cell antibodies), and adrenergic drug resistance (including adrenergic drug resistance with asthma or cystic fibrosis) (often characterized, e.g., by beta-adreneric receptor antibodies).
Additional disorders that may have an autoimmune component that may be treated, prevented, diagnosed andlor prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin-fusion proteins of the invention include, but are not limited to, chronic active hepatitis (often .characterized, e.g., by smooth ~ muscle antibodies), .primary biliary cirrhosis (often characterized, e.g., , by mitochondria IS antibodies), other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-MI (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g.; by myocardial antibodies), urticaria (often - characterized, . e.g., by IgG and IgM antibodies to IgE), atopic dermatitis (often characterized, e.g., by IgG and IgM antibodies to IgE), asthma (often characterized, e.g., by IgG and IgM antibodies to IgE), and. many' other inflammatory, granulomatous, degenerative, and atrophic disorders. ~ .
In a preferred embodiment, the autoinnmune diseases and disorders and/or , conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognbsed using for example, fusion proteins of the invention 'and/or polynucleotides encoding albumin fusion proteins of the invention. In a specific preferred embodiment, rheumatoid arthritis is treated, prevented, andlor diagnosed using fusion proteins of, the invention andlor polynucleotides encoding albumin fusion proteins of the invention.
In another specific preferred embodiment, systemic lupus erythematosus is treated, prevented, and/or diagnosed using fusion proteins of the invention,and/or polynucleotides encoding albumin fusion proteins of the ,invention. In another specific preferred embodiment, idiopathic thrombocytopenia purpura is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. ~ .
In another specific preferred embodiment IgA nephropathy is treated, prevented;

and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used' as a immunosuppressive agent(s). ' .
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin .
fusion proteins of the invention may be useful in treating, preventing, prognosing, and/or diagnosing diseases, disorders, and/or conditions of hematopoietic cells.
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, andlor conditions associated with a decrease in certain (or many) types hematopoietic cells, including but not limited to, leukopenia; neutropenia, ~ anemia, and thrombocytopenia.
Alternatively, fusion proteins of the invention and/_or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, andlor conditions associated with an increase in certain (or many) types , of hematopoietic Bells, including but not limited to, histiocytosis.
Allergic reactions and conditions, such as asthma (particularly allergic asthma) or other, respiratory problems, may also be treated, prevented, diagnosed and/or prognosed , using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Moreover, these .molecules can be used to treat, prevent, prognose, and/or diagnose anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility: .
Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used ~to treat, prevent, diagnose and/or prognose IgE-mediated allergic reactions. Such .allergic reactions include, but are not Iimited,to, asthma, rhinitis, and eczema. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate IgE concentrations in vitro or in vivo.
Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention have uses in the diagnosis, prognosis, prevention, and/or treatment of inflammatory conditions. For example, since fusion proteins of the invention and/or polynucleotides.encoding albumin fusion proteins of the invention may inhibit the activation, proliferation and/or differentiation of cells involved in an inflammatory response, these molecules can be used to prevent and/or treat chronic and acute inflammatory conditions. Such inflammatory . conditions include, but are not limited to, for example, inflammation associated with infection (e.g., septic shock, sepsis, or systemic inflammatory response syndrome), ischemia-reperfusion injury, endotoxin lethality, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, over production of cytokines (e.g., TNF or IL-l.), respiratory disorders (e.g., asthma and allergy); gastrointestinal disorders (e, g., IO inflammatory bowel disease); cancers (e.g., gastric, ovarian, lung, bladder, Liver, arid breast); CNS , disorders (e.g., multiple sclerosis; ischemic brain injury and/or stroke, traumatic brain injury, neurodegenerative , disorders (e.~., Parkinson's disease and Alzheimer's disease); AIDS-related dementia; and prion disease);
cardiovascular disorders (e.g., atherosclerosis, myocarditis, cardiovascular disease, and cardiopulmonary bypass IS complications); as wel'I as many additional diseases, conditions, and disorders that are characterized by inflammation (e.g., hepatitis, rheumatoid arthritis, gout, trauma, pancreatitis, sarcoidosis, dermatitis, renal ischemia-reperfusion injury, Grave's disease, systemic lupus erythematosus, diabetes mellitus, and allogenic transplant rejection).
Because inflammation is a fundamental defense mechanism, inflammatory disorders 20 can effect virtually any tissue of the body: Accordingly, fusion proteins , of the . invention and/or polynucleotides encoding albumin fusion proteins, of the invention, have uses in the treatment of tissue-specific inflammatory disorders, including, but not limited to, adrenalitis, alveolitis, angiocholecystitis, appendicitis, balanitis, blepharitis, bronchitis, bursitis, ~carditis, cellulitis, cervicitis, cholecystitis~ chorditis, cochlitis, colitis,, conjunctivitis, 25 cystitis, dermatitis, diverticulitis, encephalitis, endocarditis, esophagitis, eustachitis, fibrositis, folliculitis, gastritis, gastroenteritis, gingivitis, glossitis, hepatosplenitis, keratitis, Iabyrinthitis, laryngitis, Iymphangitis, mastitis, media otitis, meningitis, metritis, mucitis, myocarditis, myosititis, myringitis, nephritis; neuritis, orchitis, osteochondritis, otitis, pericarditis, peritendonitis, peritonitis, pharyngitis, phlebitis, poliomyelitis, prostatitis, 30 pulpitis, retinitis, rhinitis, salpingitis, scleritis; sclerochoroiditis, scrotitis, sinusitis, spondylitis, steatitis, stomatitis, synovitis, syringitis, tendonitis, tonsillitis, urethritis, and vaginitis.
In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are useful to diagnose, prognose, 35 prevent, and/or treat organ transplant rejections and graft-versus-host disease. Organ rejection occurs by host immune cell destruction of the ransplanted tissue through an immune response. Similarly, an immune response is also involved in GVHD, but, in this WO 01/79442 , PCT/USO1/11850 case, the foreign transplanted immune cells destroy the host tissues.
Polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD.
. 5 In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of ~T-cells, may be an effective therapy in preventing experimental allergic and hyperacute xenograft rejection.
In~ other embodiments, fusion ' proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are useful to diagnose, prognose, prevent, and/or treat immune complex diseases, including, but not limited to, serum sickness, post streptococcal glomerulonephritis~ polyarteritis nodosa, and immune complex induced vasculitis.
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin ~ fusion proteins of the invention can be used to treat, detect, and/or prevent infectious agents.
For example, by increasing the immune response; particularly increasing the proliferation activation and/or differentiation of B and/or T cells, infectious diseases may be treated, detected, and/or prevented. The immune response may be increased by either enhancing an existing immune response, or by initiating anew immune response.
Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also directly inhibit the infectious agent (refer to section of application listing infectious agents, etc), without necessarily eliciting an immune response.
In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a vaccine adjuvant that enhances immune responsiveness to an antigen. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance tumor-specific immune responses.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion, proteins of the invention are used as.an adjuvaiit to enhance anti-viral immune responses. ' Anti-viral immune responses that may b~e enhanced using the compositions of the invention as an adjuvant, include virus and virus associated diseases or symptoms described herein or otherwise known in the 1 art. In specific embodiments, the compositions of the' invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of:
AIDS, meningitis, Dengue, EBV, and hepatitis, (e.g., hepatitis B). In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an 194 .

WO 01/79442 PCT/USO1/11850 .
immune response to a virus, disease, or symptom selected from the group consisting of:
. HIV/AIDS, respiratory syncytial virus, Dengue, rotavirus,, Japanese 'B
encephalitis, influenza A,and B, parainfluenza, measles, cytomegalovirus, rabies, Junin, Chikungunya, Rift Valley Fever, herpes simplex, and yellow fever.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are~used as an adjuvant to enhance anti-bacterial or anti-fungal immune responses. Anti-bacterial or anti-fungal immune responses that may be enhanced using the compositions of the invention as an adjuvant, include bacteria or fungus and bacteria or fungus associated diseases or symptoms described herein or otherwise known in the art. ,In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: tetanus, Diphtheria, botulism, and meningitis type B.
In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: Vibrio cholerae, Mycobacterium leprae, Salmonella typhi, Salmonella paratyphi, Meisseria meningitidis, Stf-eptococcus pneumoniae, Group' B
streptococcus, Shigella spp., Enterotoxigenic Escherichia coli, Enterohemorrhagic E. coli, and B~rrelia burgdorferi. . ' In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-parasitic immune responses. Anti-parasitic immune responses that may be enhanced using the compositions of the invention as an adjuvant, include parasite and parasite associated diseases or symptoms described herein or otherwise known in the art. In r;
specif c embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a parasite. In another specific embodiment, the compositions of the invention are used~as an, adjuvant to enhance an immune response to Plasmodium (malaria) or Leishmania.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion. proteins of the invention may also be employed to treat infectious diseases.including silicosis, sarcoidosis, and idiopathic pulmonary fibrosis;
for example, by preventing the recruitment and activation of mononuclear phagocytes.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an antigen for the generation of antibodies to inhibit or enhance immune mediated responses against polypeptides of the invention.
In one embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are administered to an animal (e.g., mouse, rat, rabbit, hamster, guinea pig, pigs, micro-pig, chicken, camel, goat, horse, cow, sheep, dog, cat; , non-human primate, and human, most preferably human) to boost the immune system to produce increased quantities, of one or more antibodies (e.g., IgG, IgA, . IgM, and IgE), to induce higher affinity antibody production and immunoglobulin class switching (e.g., IgG, IgA, IgM, and IgE), and/or to increase an immune response.
In another specific embodiment, albumin fusion proteins of the invention .
and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell responsiveness to pathogens.
I0 In.another specifc embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an activator of T cells.
In another specific embodiment, albumin fusion proteins of 'the invention .and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent that elevates the immune status of an individual prior to their receipt of immunosuppressive therapies:
In another specific embodiment, albumin fusion proteins' ,of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as ari agent to induce higher affinity antibodies.
In another specific embodiment, albumin fusion proteins .of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to increase serum immunoglobulin concentrations.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as .an agent to accelerate recovery of immunocompromised individuals.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among aged populations and/or neonates.
In another specific embodiment, albumin fusion proteins ~of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an. immune system-enhancer prior to, during, or after bone marrow transplant and/or other transplants (e.g., allogeneic or xenogeneic organ transplantation). With respect to transplantation, compositions of the invention may be administered prior to, concomitant with, and/or after transplantation. In a specific embodiment, compositions of the invention are administered after transplantation, prior to the beginning of recovery of T-cell populations. In another specific embodiment, compositions of the invention are first administered after transplantation after the beginning of recovery of T cell populations, but prior to full recovery of B cell populations. . , In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having an acquired loss of B cell function.
Conditions resulting in an acquired loss of B cell function that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, HIV
Infection, AIDS, bone marrow transplant, and B cell chronic lymphocytic leukemia (CLL).
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of, the invention are used as an agent to boost immunoresponsiveness among individuals having a temporary immune deficiency.
Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include,lbut are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, and recovery from surgery.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a regulator of antigen presentation by monocytes, dendritic cells, and/or B-cells. In one embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding 'albumin fusion ' proteins of the invention enhance antigen presentation or antagonizes antigen presentation in vitro or in vivo. Moreover, in related embodiments, this enhancement or antagonism of antigen presentation may be useful as an anti-tumor treatment or to modulate the immune system.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to direct an individual's immune system towards development of a humoral response (i.e.
TH2) as opposed to a TH1 cellular response.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of tlie, invention are used as a means to induce tumor proliferation and thus make it more susceptible to anti-neoplastic agents. For example, multiple myeloma is a slowly dividing disease and is thus refractory to virtually all anti-neoplastic regimens. If these cells were forced to proliferate more rapidly their susceptibility profile would likely change. .
. In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell production in. pathologies such as AIDS, chronic lymphocyte disorder andlor Common Variable Immunodificiency.
In another specific embodiment, albumin fusion proteins . of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for generation and/or regeneration of lymphoid tissues following surgery, trauma or genetic defect. In another specific embodiment, albumin fusion proteins of the invention and/or - polynucleotides encoding albumin fusion proteins of the invention are used in the pretreatment of bone marrow samples prior to transplant.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a gene-based therapy for genetically inherited disorders ~ resulting in immuno incompetence/immunodeficiency such as observed among SCID patients.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of activating monocytes/macrophages to defend against parasitic diseases that effect monocytes such as Leishmania.
In another specific embodiment,' albumin fuaiorl proteins of the 'invention and/or polynucleotides encoding albumin fusion proteins of. the invention are used as a means of regulating secreted cytokines that are elicited by polypeptides of the invention.
In another embodiment, albumin fusion . proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in 'one or more of the applications decribed herein, as they may apply to veterinary medicine.
. In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of blocking various aspects of immune responses to foreign agents or self.
Examples of diseases or conditions in which blocking of certain aspects of immune responses may be desired include autoimmune disorders such ~as lupus, and arthritis, as well as immunoresponsiveness to skin allergies, inflammation, bowel . disease, injury and diseasesldisorders associated with pathogens.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for preventing the B cell proliferation and Ig secretion associated with autoiminune diseases such as idiopathic thrombocytopenic purpura, systemic lupus erythematosus and multiple sclerosis.
In another specific embodiment, polypeptides, antibodies, polynucleotides and/or agonists or antagonists of the present fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention invention are used as a inhibitor of B

and/or T cell migration in endothelial cells. This activity disrupts tissue architecture or cognate responses and is useful, for example in disrupting immune responses, and blocking sepsis.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for chronic hypergammaglobulinemia evident in such diseases as monoclonal gammopathy of undetermined significance (MGUS), Waldenstrom's disease, related idiopathic monoclonal gammopathies, and plasrriacytomas.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusiom proteins of the invention may be employed for instance to inhibit polypeptide chemotaxis and activation of macrophages and their . precursors, and of neutrophils, basophils, B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T cells and natural killer cells, in certain autoimmune and chronic inflammatory and infective diseases. Examples of autoimmune diseases are described herein and include multiple sclerosis, and insulin-dependent diabetes.
The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat idiopathic hyper-eosinaphilic syndrome by, for example, preventing eosinophil production and migration.
In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides~encoding albumin fusion proteins of the invention are used to enhance or inhibit complement mediated cell lysis.
. In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit antibody dependent cellular cytotoxicity.
~ In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed for treating atherosclerosis, for example, by preventing monocyte infiltration in the artery wall.
In another specific embodiment, albumin fusion proteins of the invention and/or ~ ,polynucleotides encoding albumin fusion proteins of the invention may be employed to treat adult respiratory distress syndrome CARDS).
In another specific embodiment, albumin fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention may be useful for stimulating wound and tissue repair, stimulating angiogenesis, and/or stimulating the repair of vascular or lymphatic diseases or disorders. Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to stimulate the regeneration of mucosal surfaces.

In a specific embodiment, albumin fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, treat, and/or prevent a disorder characterized by primary or acquired immunodeficiency, deficient serum immunoglobulin production, recurrent infections, and/or immune system dysfunction. Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat ~or prevent infections of the joints, bones, skin, and/or parotid glands, blood-borne infections (e.g., sepsis, meningitis,.septic arthritis, and/or osteomyelitis), autoimmune diseases (e.g., those disclosed herein), inflammatory disorders, and malignancies, and/or any disease or disorder or condition associated with these infections, diseases, disorders and/or malignancies) including, but not limited to, CVID, other primary immune deficiencies, HIV
disease, CLL, recurrent bronchitis, sinusitis, otitis media, conjunctivitis, pneumonia, hepatitis, meningitis, herpes zoster (e.g.; severe herpes zoster), and/or pneumocystis carnii.
Other diseases and disorders that may be prevented, diagnosed, prognosed, arid7or treated with fusion ~ proteins of , the invention and/or polynucleotides. encoding albumin- fusion proteins of the invention include, but are not limited to, HIV infection, HTLV-BLV
infection, lymphopenia, phagocyte bactericidal dysfunction anemia, thrombocytopenia, and hemoglobinuria.
' In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat, and/or diagnose an individual having common variable immunodeficiency disease ("CVID"; also known as "acquired agammaglobulinemia" and "acquired hypogammaglobulinemia") or a subset of this disease.
In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to diagnose, prognose, prevent, and/or treat cancers or neoplasms including immune cell or immune tissue-related cancers or neoplasms. Examples of cancers or neoplasms that may be prevented, diagnosed, or treated by fusion proteins of the invention .and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, acute myelogenous leukemia, chronic myelogenous leukemia Hodgkin's disease, non-Hodgkin's lymphoma, acute Iymphocytic anemia (ALL) Chronic lymphocyte leukemia, plasmacytomas, multiple myeloma, Burkitt's lymphoma, EBV-transformed diseases, and/or ' diseases and disorders described in the section entitled "Hyperproliferative Disorders"
elsewhere herein.
~In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapyfor decreasing cellular proliferation of Large B-cell Lymphomas.
20.0 In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of , decreasing the involvement of B cells and Ig associated with Chronic Myelogei~ous Leukemia.
5_ In specific embodiments, the compositions of the invention are used as an agent to boost immunoresponsiveness among B cell immunodeficient individuals, such as, for example, an individual who has undergone a partial or complete splenectomy.
Blood-Related Disorders The albumin ~ fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention may be used to modulatef hemostatic (the stopping of bleeding) or thrombolytic (clot dissolving) activity. for example, by increasing hemostatic or thrombolytic activity, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to treat or prevent blood coagulation diseases, disorders, and/or conditions (e.g., afibrinogenemia, factor deficiencies, hemophilia), blood platelet diseases, disorders, and/or conditions (e.g., thrombocytopenia), or 'wounds resulting from trauma, surgery, or other causes.
Alternatively, fusion proteins of ~ the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can decrease hemostatic or thrombolytic activity could be used to inhibit or dissolve clotting. These molecules could be important in the treatment or prevention of heart attacks (infarction), strokes, or scarring.
In specific embodiments, the albumin fusion proteins of the invention and/or .
polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, diagnose, prognose, and/or treat thrombosis, arterial thrombosis, venous thrombosis, . thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina. In specific embodiments, the albumin fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention may be used for the prevention of occulsion of saphenous grafts, for reducing .the risk of periprocedural thrombosis as might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonrheumatic atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mural valves disease. Other uses for the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, the ' prevention of occlusions in extrcorporeal devices (e.g., intravascular canulas, vascular .
access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines).

In another embodiment, albumin fusion proteins of ~ the invention and/or -polynucleotides encoding albumin fusion proteins of the invention, may be used to prevent, diagnose, prognose, and/or treat diseases and' disorders of the blood and/or blood forming organs associated with the tissues) in which the polypeptide of the invention is expressed.
The fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hematopoietic .
activity (the formation of blood cells). For example, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to increase the quantity. of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets. The ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of anemias and leukopenias described below. Alternatively, the albumin fusion proteins, of the invention and/or polynucleotides encoding albumin fusion proteins of.the invention may be used to decrease the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets.. The ability to decrease the quantity of blood cells' or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of leukocytoses, such as, for example-eosinophilia.
The fusion proteins of 'the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, treat, or diagnose blood dyscrasia.
Anemias are conditions in which the number of red blood cells or amount of hemoglobin (the protein that carries oxygen) in them is below normal. Anemia may be caused by excessive bleeding, decreased red blood cell production, or increased red blood cell destruction (hemolysis). The albumin fusion . proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias. Anemias that may be treated prevented or diagnosed by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include iron deficiency anemia, hypochromic anemia, microcytic anemia, chlorosis, hereditary siderob;astic anemia, idiopathic acquired sideroblastic anemia, red cell aplasia, megaloblastic anemia (e.g., pernicious anemia, (vitamin B 12 def ciency), and folic acid deficiency anemia), aplastic anemia, hemolytic anemias (e.g., autoimmune helolytic anemia, microangiopathic hemolytic anemia, and paroxysmal nocturnal hemogIobinuria). The albumin fusion proteins of the~invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with diseases including but not limited ~to, anemias associated with systemic lupus erythematosus; cancers, lymphomas, chronic renal disease, and enlarged spleens. The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating,.
preventing, and/or diagnosing anemias arising from drug treatments such as anemias associated with methyldopa, dapsone, and/or sulfadrugs. Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with abnormal red blood cell architecture including but not limited to, hereditary spherocytosis, hereditary elliptocytosis, glucose-6-phosphate dehydrogenase deficiency, and sickle cell anemia.
The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing hemoglobin abnormalities, (e.g., those associated with sickle cell anemia, hemoglobin C disease, hemoglobin S-C disease, and hemoglobin E disease).
Additionally, .the albumin fusion proteins of the invention and/or polynucleotides~encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, andlor treating thalassemias, including, but not limited to, major and minor forms of alpha-thalassemia and beta-thalassemia..
In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating bleeding disorders including, but not limited to, thrombocytopenia (e.g., idiopathic thrombocytopenic purpura, and thrombotic thrombocytopenic purpura), Von Willebrand's disease, hereditary platelet disorders (e.g., storage pool disease such as ~Chediak-Higashi and Hermansky-Pudlak syndromes, thromboxane A2' dysfunction, thromboasthenia, and Bernard-Soulier syndrome), hemolytic-uremic syndrome, hemophelias such as hemophelia A or Factor VII
deficiency and Christmas disease or Factor IX deficiency, Hereditary Hemorhhagic Telangiectsia, also known as Rendu-Osler-Weber syndrome, allergic purpura (Henoch Schonlein purpura) and disseminated intravascular coagulation.
The effect of the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention on the clotting time of blood may be monitored using any of the clotting tests known in the art including, but not limited to, whole blood partial thromboplastin time (PTT), the activated partial thromboplastin time (aP'TT), tle activated clotting time (ACT'), the recalcified activated clotting time, or the Lee-White Clotting time.
Several diseases and a variety of drugs can cause platelet dysfunction. Thus, in a specific embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating acquired platelet dysfunction such as platelet dysfunction 203 . ' accompanying kidney failure, leukemia, multiple myeloma, cirrhosis of the liver, and systemic lupus erythematosus as well as platelet dysfunction associated with drug treatments, including treatment with aspirin, ticlopidine, nonsteroidal anti-inflammatory drugs (used for arthritis, pain, and sprains), and penicillin in high doses.
In another embodiment, the albumin fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders characterized by or associated with increased or decreased numbers of white blood cells.
Leukopenia occurs when the number of white blood cells decreases below normal. Leukopenias include, but are not limited to, neutropenia and lymphocytopenia. An increase in the number of white blood cells compared to normal is known as leukocytosis. The body generates increased . numbers of white blood cells during infection. Thus, leukocytosis may simply be a normal physiological parameter that reflects infection: Alternatively, leukocytosis may be an indicator of injury or other disease such as cancer. Leokocytoses, include but are not , limited to, eosinophilia, and accumulations of macrophages. In specific embodiments; the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in ,diagnosing, prognosing, preventing, .and/or treating leukopenia. In other specific embodiments, the albumin fusion proteins, of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukocytosis.
Leukopenia may be a generalized decreased in all types of white blood cells, or may be a specific depletion of particular types of white blood cells. Thus, in specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating decreases in neutrophil numbers, known as neutropenia.
Neutropenias that may be diagnosed, ptognosed, prevented, and/or treated by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, infantile genetic agranulocytosis, familial neutropenia, cyclic' neutropenia, neutropenias resulting from or associated with dietary deficiencies (e.g., vitamin B 12 deficiency or folic acid deficiency), neutropenias resulting from or associated with drug. treatments (e.g.,~ antibiotic regimens such .as penicillin treatment, sulfonamide treatment; anticoagulant treatment, anticonvulsant drugs, anti-thyroid drugs, and'cancer chemotherapy), and neutropenias resulting from increased neutrophil destruction that may occur in association with some bacterial or viral infections, allergic disorders, autoimmune diseases, conditions in which an individual has an enlarged spleen (e.g., Felty syndrome, malaria and sarcoidosis), and some drug treatment regimens.
The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating lymphocytopenias (decreased numbers of B and/or T
lymphocytes), including, but not limited to, lymphocytopenias resulting from or associated -with stress, drug treatments (e.g., drug treatment with corticosteroids, cancer chemotherapies, andlor radiation therapies), AIDS infection and/or other diseases such as, . for example, cancer, rheumatoid arthritis, systemic lupus erythematosus, chronic infections, some viral infections and/or hereditary disorders (e.g., DiGeorge syndrome, Wiskott Aldrich Syndome, severe combined immunodeficiency, ataxia telangiectsia).
The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with macrophage numbers and/or macrophage function including, but not limited to, Gaucher's disease, Niemann-Pick disease, Letterer-Siwe disease and Hand-Schuller-Christian disease.
In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the y invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with eosinophil numbers and/or eosinophil .function including, but not limited to, idiopathic hypereosinophilic syndrome, eosinophilia-myalgia syndrome, and Hand-Schuller-Christian disease.
In yet another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukemias and lymphomas including, but not limited to, acute lymphocytic (lymphpblastic) leukemia (ALL), acute myeloid (myelocytic, myelogenous, myeloblastic, or myelomonocytic) leukemia, chronic ..lymphocytic leukemia (e.g., B cell leukemias, T cell-leukemias, Sezary syndrome, and . Hairy cell leukenia), chronic myelocytic (myeloid, myelogenous, or granulocytic) leukemia, Hodgkin's lymphoma, non-hodgkin's lymphoma, Surkitt's lymphoma; and mycosis fungoides. , In other embodiments, the albumin fusion proteins of the invention and/or 30. polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders of plasma cells ' including, but not limited to, plasma cell dyscrasias, monoclonal gammaopathies, monoclonal ~gammopathies of undetermined significance, multiple myeloma, macroglobulinemia, Waldenstrom's macroglobulinemia, cryoglobulinemia, and Raynaud's phenomenon. _ ~ .
In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing myeloproliferative disorders, including but not limited to, polycythemia vera, relative polycythemia, secondary polycythemia, myelofibrosis, acute myelofibrosis, agnogenic myelod metaplasia, thrombocythemia, (including both primary and seconday thrombocythemia) and chronic myelocytic leukemia.
In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as a treatment prior to surgery, to increase blood cell production.
In other embodiments, the ~ albumin fusion proteins of .the ,invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to enhance the migration, phagocytosis, superoxide production, antibody dependent cellular cytotoxicity of neutrophils, eosionophils and macrophages.
In other embodiments, the - albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to stem cells pheresis. In another specific embodiment, the albumin fusion proteins of the , invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to platelet pheresis.
In other embodiments, the albumin fusion proteins of , the invention and/or .
polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase cytokine production.
- In other embodiments, the albumin fusion ~ proteins 'of the invention andlor _ polynucleotides encoding albumin fusion proteins of the invention may .be useful in preventing, diagnosing, and/or treating primary hematopoietic disorders.
Hv~erproliferative Disorders In certain embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat or detect hyperproliferative disorders, including neoplasms. Albumin fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention may inhibit the . ~ proliferation of the disorder through direct or indirect interactions.
Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may proliferate other cells which can inhibit the hyperproliferative disorder.
For example, by increasing an immune response, particularly increasing antigenic qualities of the hyperproliferative disorder or by proliferating, differentiating, or mobilizing T-cells, hyperproliferative disorders can be treated. This immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response. Alternatively, decreasing an immune response may also be a method of treating hyperproliferative disorders, such as a chemotherapeutic agent.
Examples of hyperproIiferative disorders that can be treated or detected by-fusion - proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the 5, invention include, but are not limited to neoplasms located in the: colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck; nervous (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and uxogenifal tract.
Similarly, other hyperproliferative disorders can also be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of such hyperproliferative disorders include, but are not limited to:
Acute Childhood Lymphoblastic Leukemia; Acute Lymphoblastic Leukemia, Acute Lymphocytic Leukemia, Acute Myeloid Leukemia, Adrenocortical Carcinoma, Adult (Primary) Hepatocellular Cancer, Adult (Primary) Liver Cancer, Adult Acute Lymphocytic Leukemia, Adult Acute Myeloid Leukemia, Adult Hodgkin's Disease, Adult Hodgkin's Lymphoma, Adult Lymphocytic Leukemia, Adult Non-Hodgkin's Lymphoma, Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Related Lymphoma, AIDS-Related Malignancies, Anal Cancer, Astrocytoma, Bile Duct Cancer, Bladder Cancer, Bone Cancer, Brain Stem Glioma, Brain Tumors, Breast Cancer, Cancer of the Renal Pelvis and Ureter, Central Nervous System (Primary) Lymphoma, Central Nervous System Lymphoma, Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Childhood (Primary) Hepatocellular Cancer, Childhood (Primary) Liver Cancer, Childhood Acute Lympholilastic Leukemia, Childhood Acute Myeloid Leukemia, Childhood Brain Stem Glioma, Childhood Cerebellar Astrocytoma, Childhood Cerebral Astrocytoma, Childhood Extracranial Germ Cell Tumors, Childhood Hodgkin's Disease, Childhood Hodgkin's Lymphoma, Childhood Hypothalamic and Visual Pathway Glioma, Childhood Lymphoblastic ' Leukemia, Childhood Medulloblastoma, Childhood Non-Hodgkin's Lymphoma, Childhood Pineal and Supratentorial Primitive Neuroectodermal Tumors, Childhood Primary Liver Cancer, Childhood Rhabdomyosarcoma, Childhood Soft Tissue Sarcoma, Childhood Visual Pathway and Hypothalamic Glioma, Chronic Lymphocytic Leukemia, Chronic Myelogenous Leukemia, Colon Cancer, Cutaneous T-Cell Lymphoma, Endocrine Pancreas Islet Cell Carcinoma, Endometrial ~ Cancer, Ependymoma, Epithelial Cancer, Esophageal Cancer, Ewing's Sarcoma and Related Tumors, Exocrine Pancreatic Cancer, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Eye Cancer, Female Breast Cancer, Gaucher's Disease, Gallbladder Cancer, Gastric Cancer, Gastrointestinal Carcinoid Tumor; Gastrointestinal Tumors, Germ Cell Tumors, Gestational Trophoblastic Tumor, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular Cancer, Hodgkin's Disease, Hodgkin's Lymphoma, Hypergammaglobulinemia, Hypopharyngeal Cancer, Intestinal Cancers, Intraocular Melanoma, Islet Cell Carcinoma, Islet Cell Pancreatic Cancer, Kaposi's Sarcoma, Kidney S ~ Cancer, Laryngeal Cancer, Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer, Lymphoproliferative Disorders, Macroglobulinemia, Male Breast Cancer, Malignant Mesothelioma, Malignant Thymoma, Medulloblastoma, Melanoma, Mesothelioma, Metastatie Occult Primary Squamous Neck Cancer, Metastatic Primary Squamous Neck Cancer, Metastatie Squamous Neck-Cancer, Multiple Myeloma, Multiple Myeloma/Plasma Cell Neoplasm, Myelodysplastic Syndrome, Myelogenous Leukemia, Myeloid Leukemia, Myeloproliferative Disorders, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin's Lymphoma During Pregnancy, Nonmelanoma Skin Cancer, Non-Small Cell Lung Cancer, Occult Primary Metastatic Squamous Neck Cancer, Oropharyngeal ~ Cancer, Osteo-/Malignant . Fibrous Sarcoma, Osteosarcoma/lVlalignant Fibrous Histiocytoma, Osteosarcoma/Malignant Fibrous Histiocytoma of Bone, Ovarian Epithelial Cancer, Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor, Pancreatic Cancer, Paraproteinemias, Purpura, Parathyroid.
Cancer, Penile Cancer, Pheochromocytoma, Pituitary Tumor, . Plasma Cell Neoplasm/Multiple Myeloma, Primary Central Nervous System Lymphoma, Primary Liver ~ Cancer, ~ Prostate Cancer, Rectal Cancer,' Renal Cell Cancer, Renal Pelvis and Ureter Cancer, Retinoblastoma, Rhabdomyosarcoma, Salivary Gland Cancer, Sarcoidosis Sarcomas, Sezary 'Syndrome, Skin Cancer, Small Cell Lung Cancer, Small Intestine Cancer, Soft Tissue Sarcoma, Squamous Neck Cancer, Stomach Cancer, Supratentorial - Primitive Neuroectodermal and Pineal Tumors, T-Cell Lymphoma, Testicular Cancer, Thymoma, Thyroid Cancer, Transitional Cell Cancer of ' the Renal Pelvis and Ureter, Transitional Renal Pelvis and Ureter Cancer, Trophoblastic Tumors, Ureter and Renal Pelvis Cell Cancer, Urethral Cancer, Uterine Cancer, Uterine Sarcoma, Vaginal Cancer, Visual Pathway and Hypothalamic Glioma, Vulvar Cancer, Waldenstrom's Macroglobulinemia, Wilms' Tumor, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above. ' In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, prevent; and/or treat premalignant conditions and to prevent progression to a neoplastic or malignant state, including but not limited to those disorders described 'above.
Such uses are indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non-neoplastic cell growth consisting of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for review of such WO 01/79442 . PCT/USO1/11850 abnormal growth conditions, see Robbins. and Angell, 1976, Basic Pathology, 2d Ed., W.
B. Saunders Co., Philadelphia, pp. 6~-79.) Hyperplasia is a form of controlled cell proliferation, involving an increase in cell number in a tissue or organ, without significant alteration in structure or function.
Hyperplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, angiofollicular mediastinal lymph node hyperplasia, angiolymphoid hyperplasia with eosinophilia, atypical melanocytic hyperplasia, basal cell hyperplasia, benign giant lymph node hyperplasia, cementum hyperplasia, congenital adrenal hyperplasia, congenital sebaceous hyperplasia, cystic hyperplasia, cystic hyperplasia of the breast, denture hyperplasia, ductal hyperplasia, endometrial hyperplasia, fibromuscular hyperplasia, focal epithelial hyperplasia, gingival hyperplasia, inflammatory fibrous hyperplasia, inflammatory papillary hyperplasia, intravascular papillary endothelial hyperplasia, nodular hyperplasia of prostate, nodular regenerative hyperplasia, pseudoepitheliomatous hyperplasia, senile sebaceous hyperplasia, and verrucous hyperplasia.
Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell. Metaplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, agnogenic myeloid metaplasia, apocrine metaplasia, atypical metaplasia, autoparenchymatous metaplasia, connective tissue metaplasia, epithelial metaplasia, intestinal metaplasia, metaplastic anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia, primary myeloid metaplasia, secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of amnion, and symptomatic myeloid metaplasia.
Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-neoplastic cell growth, involving a loss in individual.cell uniformity and in the architectural orientation of cells. Dysplastic cells often have abnormally large, deeply stained nuclei, and exhibit pleomorphism. Dysplasia characteristically occurs where there exists chronic irritation or inflammation. Dysplastic disorders which can be diagnosed; prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia; encephalo-ophthalmic dysplasia, dysplasia epiphysialis .
hemimelia, dysplasia epiphysialis multiplex, dysplasia epiphysialis punctata, epithelial dysplasia, faciodigitogenital dyspIasia, familial fibrous. dysplasia of jaws, familial white folded dysplasia, fibromuscular dysplasia, fibrous dysplasia of bone, florid osseous dysplasia, hereditary renal-retinal dysplasia; hidrotic ectodermal dysplasia, hypohidrotic ectodermal dysplasia, lymphopenic thymic dysplasia, mammary dysplasia, mandibulofacial dysplasia, . metaphysial dysplasia, Mondini dysplasia, monostotic fibrous dysplasia, ~mucoepithelial dysplasia, multiple epiphysial dysplasia, oculoauriculovertebral dysplasia, oculodentodigital dysplasia, oculovertebral dysplasia, : odontogenic dysplasia, ophthalmomandibulomelic dysplasia, periapical cemental dysplasia, polyostotic fibrous dysplasia, pseudoachondroplastic spondyloepiphysial dysplasia, retinal dysplasia, septo-optic dysplasia, spondyloepiphysial dysplasia, and ventriculoradial dysplasia.
Additional. pre-neoplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin , fusion proteins of the invention include, but are not limited to, benign dysproliferative disorders (e.g., benign tumors, . fibrocystic conditions; tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia), leukoplakia, keratoses, Bowen's disease, _ Farmer's Skin, solar cheilitis, and solar keratosis. ' In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to diagnose and/or prognose disorders' associated with the tissues) iW vhich the polypeptide of the invention is expressed. .
In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat cancers and neoplasms, including, but not limited to, those described herein. In a further preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat acute myelogenous leukemia.
3.0 Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may affect apoptosis, and therefore, would be useful in treating a number of diseases associated with increased cell survival or the inhibition of apoptosis. For example, diseases associated with increased cell survival or the inhibition of apoptosis that could be diagnosed, prognosed, prevented, and/or treated by polynucleotides, polypeptides, and/or agonists or _antagonists~ of the invention, include cancers ~ (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, ~ neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders such as, multiple sclerosis, Sjogren's syndrome, Hashimoto'~s thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related ' glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection.
In preferred embodiments, fusion proteins of the invention and/or .
polynucleotides encoding albumin fusion proteins of the invention are used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above.
Additional diseases or conditions associated with increased cell survival that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myeIocytic (granuIocytic) leukemia 2~0 and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and . carcinomas such , as fibrosarcoma, myxosarcoma, liposarcoma;
chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon ' carcinoma, pancreatic cancer, breast - cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile . duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, . testicular tumor, lung _ carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, emangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
Diseases associated with increased apoptosis that could be diagnosed, prognosed, prevented; and/or treated by fusion proteins of the invention andlor polynucleotides ' ~ 211 encoding albumin fusion proteins of the invention, include AIDS;
neurodegenerative .
disorders (such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, cerebellar degeneration and brain tumor or prior associated disease);
autoimmune disorders' (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury .
(such as that caused by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/ieperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
Hyperproliferative diseases and/or disorders that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins. of the invention, include, but are not limited to, neoplasms located in. the liver, abdomen, bone, breast, digestive system, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, headband neck, nervous system (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract.
Similarly, other hyperproliferative disorders can also be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of such hyperproliferative disorders include, but are not limited to: hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias,, purpura, sarcoidosis, Sezary Syndrome, .Waldenstron's macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
Another preferred embodiment utilizes polynucleotides encoding albunnin fusion proteins of the invention to inhibit aberrant cellular division, by gene therapy using the present invention, and/or protein fusions or fragments thereof.
Thus, the present invention provides a method for treating cell proliferative disorders . by inserting into an abnormally proliferating cell a polynucleotide encoding an albumin fusion _ protein of the present invention, wherein said polynucleotide represses said expression.
Another embodiment of the present invention provides a method of treating cell-proliferative disorders in individuals comprising administration of one or more active gene copies of the present invention to an abnormally proliferating cell or cells.
In a preferred embodiment, polynucleotides of the present invention is a DNA construct comprising a recombinant expression vector effective in expressing a DNA sequence encoding said polynucleotides. In another preferred embodiment of the present invention, the DNA
construct encoding the fusion protein of the present invention is inserted into cells to be treated utilizing a retrovirus, or more preferably an adenoviral vector (See G
J. Nabel, et.
al., PNAS 1999 96: 324-326, which is hereby incorporated by reference). In a most preferred embodiment, the viral vector is defective and will not transform non-proliferating cells, only proliferating cells. Moreover, in a preferred embodiment, the polynucleotides of the present invention inserted into proliferating cells either alone, or in combination with or fused to other polynucleotides, can then be modulated via an external stimulus (i.e.
magnetic, specific small molecule, chemical, or drug administration, etc.), which acts upon the promoter upstream of said polynucleotides to induce expression of the encoded protein product. As such the beneficial therapeutic affect of the present invention may be expressly modulated (i.e. to increase, decrease, or inhibit expression of the present invention) based upon said external stimulus.
Polynucleotides of the present invention may be useful in repressing expression of oncogenic genes or antigens. By "repressing expression of the .oncogenic genes " is intended the suppression of the transcription of the gene, the degradation of the gene .transcript (pre-message RNA), the inhibition of splicing, the destruction of the messenger RNA, the prevention of the post-translational modifications of the protein, the destruction of the protein, or the inhibition of the normal function of the protein.
For local administration to abnormally proliferating cells, polynucleotides of the present invention may be administered by any method known to those of skill in the art including, but not limited to transfection, electroporation, mieroinjection of cells, or in vehicles such as liposomes, lipofectin, or as naked polynucleotides, or any other method described throughout the specification. The polynucleotide of the present invention may be delivered by known gene delivery systems such a~s, but not limited to, retroviral vectors (Gilboa, J. Virology 44:845 (1982); Hocke, Nature 320:275 (19$6); Wilson, et al., Proc.
Natl. Acad. Sci. U.S.A. 85:3014), vaccinia virus system (Chakrabarty et al., Mol. Cell Biol. 5:3403 (1985) or other efficient DNA delivery systems (Yates.et al., Nature 313:812 ( 1985)) known to those skilled in the art. These references are exemplary only and are hereby incorporated by reference. In order to specif cally deliver or transfect cells which are abnormally proliferating and spare non-dividing cells, it is preferable to, utilize a retrovirus, or adenoviraI (as described in the art and elsewhere herein) delivery system known to those of skill in the art. Since host DNA replication is required for retroviral DNA
to integrate and, the retrovirus will be unable to self replicate due to the lack of the retrovirus genes needed 35' for its life cycle. Utilizing such a retroviral delivery system for polynucleotides of the present invention will target said gene and constructs to abnormally proliferating cells and _ will spare the non-dividing riormal cells.

The polynucleotides of the present invention may be delivered directly to cell proliferative disorder/disease sites in internal organs, body cavities. and the like by use of 1 imaging devices used to guide an injecting needle directly to the disease site. The polynucleotides of the present invention may also be administered to disease sites at the time of surgical intervention.
By "cell proliferative disease" is meant any human or animal disease or disorder, affecting any one or any combination of organs, cavities, or body parts, which is characterized by single or multiple local abnormal proliferations of cells, groups of cells, or tissues, whether benign or malignant.
Any amount of the polynucleotides of the present, invention may be administered as long as it has a biologically inhibiting effect on the proliferation of the treated cells.
Moreover, it is possible to administer more than one, of the polynucleotide of the present invention simultaneously to the same site. . By "biologically inhibiting" is meant partial or total growth inhibition as.well as decreases in the rate of proliferation or growth of the cells.
The biologically inhibitory dose may be determined by assessing the effects of the polynucleotides of the present invention on target malignant or abnormally proliferating cell growth in tissue culture, tumor growth in animals and cell cultures, or any other method known to one of ordinary skill in the art.
Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention bf the present invention are useful in inhibiting the angiogenesis of proliferative cells or tissues, either alone, as a protein fusion, or in combination with other polypeptides directly or indirectly, as described elsewhere herein. In a most preferred embodiment, said anti-angiogenesis effect may be achieved indirectly, for example, through the inhibition of hematopoietic, tumor-specific cells, such as . tumor associated macrophages (See Joseph IB, et al. J Natl Cancer Inst, 90(21):1648-53 (1998), which is hereby incorporated by reference). ;
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in inhibiting proliferative cells or tissues through the induction of apoptosis. These fusion protieins and/or polynucleotides may act either =directly, or indirectly to induce apoptosis of proliferative cells and tissues, for example in the activation of a death-domain receptor, such as tumor necrosis factor (TNF) receptor-1, CD95 (Fas/APO-1), TNF-receptor-related apoptosis-mediated protein (TRAMP) and TNF-related apoptosis-inducing ligand (TRAIL) receptor-1 and -2 (See Schulze-Osthoff K, et.al., Eur J Biochem_ 254(3):439-59 (1998), which is hereby incorporated by reference). Moreover, in another preferred embodiment of the present invention,' these fusion proteins and/or, polynucleotides, may induce apoptosis through other mechanisms, such as in the activation of other proteins which will activate apoptosis, or through stimulating the expression of these proteins, either alone or in combination with small molecule drugs or adjuviants, such as apoptonin; galectins, thioredoxins, anti-inflammatory proteins (See for example, Mutat Res 400(1-2):447-55 (1998), Med Hypotheses.50(5):423-33 (1998), Chem Biol Interact. Apr 24;111-112:23-34 (1998), J
Mol~Med.76(6,):402-12 (1998), Int J Tissue React;20(1):3-15 (1998), which are all hereby incorporated by reference).
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are useful in inhibiting the metastasis of proliferative cells or tissues. Inhibition may occur as a direct result of administering these albumin fusion IO proteins andlor poIynucleotides, or indirectly, such as activating the expression of proteins known to inhibit metastasis, for example alpha 4 integrins, (See, e.g., Curr Top Microbiol Immunol 1998;231:125-41, which is hereby incorporated by reference). Such thereapeutic affects of the present invention may be achieved either alone, or in combination with small molecule drugs or adjuvants.
In~ another embodiment, the invention provides a method of delivering compositions containing the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to targeted cells expressing the a polypeptide bound by, that binds to, or associates with an albumin fuison protein of the invention. Albumin . fusion proteins of the invention may be associated with with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via,hydrophobic, hydrophilic, ionic and/or covalent interactions.
Albumin fusion proteins of the invention are useful in enhancing the immunogenicity and/or antigenicity of proliferating cells or tissues, either directly, such as would occur if the albumin fusion proteins of the invention 'vaccinated' the immune response to respond to proliferative antigens and immunogens, or. indirectly, such as in activating the expression of proteins known to enhance the immune response (e.g. chemokines), to said antigens and immunogens.
Renal Disorders Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognose disorders of the renal system. Renal disorders which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, kidney failure, nephritis, blood vessel disorders of kidney, metabolic and congenital kidney disorders, urinary disorders of the kidney, autoimmune disorders, sclerosis and necrosis, electrolyte imbalance, and kidney cancers.

WO 01/79442 ~ PCT/USO1/11850 Kidney diseases which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, acute -kidney failure, chronic kidney failure, atheroembolic renal failure, end-stage renal disease, inflammatory diseases . of the kidney (e.g., acute glomerulonephritis, postinfectious glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic syndrome, membranous glomerulonephritis, . familial nephrotic syndrome, membranoproliferative glomerulonephritis I and II, mesangial proliferative glomerulonephritis, chronic glomerulonephritis, acute tubulointerstitial nephritis, chronic tubulointerstitial nephritis, acute post-streptococcal -glomerulonephritis (PSGN), pyelonephritis, lupus nephritis, chronic nephritis, interstitial nephritis, and post streptococcal glomerulonephritis), blood vessel disorders of the kidneys (e.g., kidney infarction, atheroembolic kidney disease, cortical necrosis, malignant nephrosclerosis, renal vein thrombosis, renal underperfusion, renal retinopathy, renal ischemia-reperfusion, renal artery embolism, and renal artery stenosis), and kidney disorders resulting form urinary . tract disease (e:g., pyelonephritis, hydronephrosis, urolithiasis (renal lithiasis, nephrolithiasis), reflux nephropathy, urinary tract infections, urinary retention, and acute or chronic unilateral obstructive uropathy.) In addition, compositions of the invention can be used to diagnose, prognose, prevent, and/or treat metabolic and congenital disorders of the kidney (e.g., uremia, renal amyloidosis, renal osteodystrophy, renal tubular acidosis, renal glycosuria, nephrogenic diabetes irisipidus, cystinuria, Fanconi's syndrome, renal fibrocystic osteosis (renal rickets), Hartnup disease, Bartter's syndrome, Liddle's syndrome, polycystic kidney .
disease, medullary cystic disease, medullary sponge kidney, Alport's syndrome, nail-patella .
syndrome, congenital nephrotic syndrome, CRUSH syndrome, horseshoe kidney, diabetic nephropathy, nephrogenic diabetes insipidus, analgesic nephropathy, kidney stones, and membranous nephropathy), and autoimmune disorders of the kidney (e.g., systemic lupus erythematosus (SLE), Goodpasture syndrome, IgA iiephropathy, and IgM mesangial proliferative ~glomerulonephritis).
Compositions of the invention can also be used to diagnose, prognose, prevent, and/or treat sclerotic or decrotic disorders of the kidney (e.g., glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis (FSGS), necrotizing glomerulonephritis, , and 1 renal papillary necrosis), cancexs of the kidney (e.g., nephroma, hypernephroma, nephroblastoma, renal cell cancer, transitional cell cancer, renal adenocarcinoma, squamous ' cell cancer, and Wilm's tumor), and electrolyte imbalances (e.g., nephrocalcinosis, pyuria, edema, hydronephritis, proteinuria, hyponatremia, hypernatremia, hypokalemia, hyperkalemia, hypocalcemia, hypercalcemia; hypophosphatemia, and hyperphosphatemia).
Compositions of the invention may be administered using. any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous c injection~~topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositoriai solid pharmaceutical formulations, decanting or topical applications during .
surgery, aerosol delivery. Such .methods are known in the art. Compositions of the invention may be administered as part of a Therapeutic, described in more detail below.
Methods of delivering polynucleotides of the invention are described in more detail herein. .
Cardiovascular Disorders .
10' Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognose cardiovascular disorders, including, but not limited to, peripheral artery disease, such as limb ischemia.
Cardiovascular disorders- include, . but are not limited to, cardiovascular abnormalities, such as arterio-arterial fistula, arteriovenous fistula, cerebral arteriovenous malformations, . congenital heart defects, pulmonary atresia, and Scimitar-Syndrome.
Congenital heart defects include, but are not limited to, aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septal defect, endocardial cushion defects, Lutembacher's Syndrome, trilogy of Fallot, ventricular heart septal defects.
Cardiovascular disorders also include, but are not. limited to, heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial),.heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy; left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, .pericardial effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculosis.
Arrhythmias include, but are not limited to, sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes Syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine Syndrome, Mahaim-type WO 01/79442 ~ ' PCT/USO1/11850 r pre-excitation sy ~ drome, WolfF Parkinson-White syndrome, sick sinus syndrome, tachycardias, and ' ventricular fibrillation. Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic functional tachycardia, sinoatrial nodal reentry tachycardia, sinus. tachycardia, Torsades de Pointer, and ventricular tachycardia.
Heart valve diseases include, but are not limited, to, aortic valve insufficiency, aortic valve stenosis, hear murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mural valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis.
Myocardial diseases include, but are not limited to, alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary. subvalvular stenosis, restrictive' cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, .Kearns Syndrome, myocardial reperfusion injury, and myocarditis.
- Myocardial ischemias include, but are not limited to, coronary disease, such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease, Klippel Trenaunay-Weber Syndrome; Sturge-Weber Syndrome, angioneurotic edema, aorkic diseases, Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial occlusive diseases, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular disorders, diabetic angiopathies, diabetic retinopathy, embolisms, thrombosis, erythromelalgia, hemorrhoids, hepatic veno occlusive ~ disease, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occlusive disease, Raynaud's disease, CREST
syndrome, retinal vein occlusion, Scimitar syndrome; superior vena cava syndrome, telangiectasia, atacia telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose veins, varicose ulcer, vasculitis, and venous insufficiency.
Aneurysms include, but are not limited to, dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
Arterial occlusive diseases include, but are not limited to, arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans.
Cerebrovascular disorders include, but are not limited to, carotid artery diseases, ~~,.--cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, - cerebral arteriovenous malformation, cerebral artery diseases, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subaraxhnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient), subclavian steal syndrome, periventricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency.
Embolisms include, but are not limited to, air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromoboembolisms. Thrombosis include, but are not limited to, coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebiti-s.
' Ischemic disorders include, but are not limited to, cerebral ischemia, ischemic colitis, compartment syndromes, ~ anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia. Vasculitis includes, but is not limited to, .
aortitis, arteritis, Behcet's Syndrome, Churg-Strauss Syndrome, mucocutaneous lymph node syndrome, thromboailgiitis obliterans, hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis. ' Albumin fusion proteins of the invention and/or polynucleotides encoding albumin .
fusion proteins of the invention may be administered rising any method known in the art, ~ ' including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulation's, decanting or topical applications during . surgery, aerosol delivery. Such methods are known in the art. Methods of delivering polynucleotides are described in more detail herein.
Respiratory Disorders Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention maybe used to treat, .prevent, diagnose, andlor prognose diseases and/or disorders of the respiratory system. ~.
Diseases and disorders of the respiratory system include, but are not limited to, nasal vestibulitis, nonallergic rhinitis (e.g., acute rhinitis, ,chronic rh~nitis, atrophic rhinitis, vasomotor rhinitis), nasal polyps, and sinusitis, juvenile angiofibromas, cancer of the nose and juvenile papillomas, vocal cord polyps, nodules (singer's nodules), contact ulcers, vocal cord paralysis, laryngoceles, pharyngitis (e.g., viral and bacterial), tonsillitis, tonsillar cellulitis, parapharyngeal abscess, laryngitis, laryngoceles, and throat~cancers (e.g., cancer of the nasopharynx, tonsil cancer, larynx cancer), lung cancer (e.g., squamous cell carcinoma, small cell (oat cell) carcinoma, large cell carcinoma, and adenocarcinoma), allergic disorders (eosinophilic pneumonia, hypersensitivity pneumonitis (e.g., extrinsic allergic alveolitis, allergic interstitial pneumonitis, organic dust pneumoconiosis, allergic bronchopulmoriary aspergillosis, asthma, Wegener's , granulomatosis (granulomatous vasculitis), Goodpasture's syndrome)), pneumonia (e.g., bacterial pneumonia (e.g., Streptococcus pneumoniae ~ (pneumoncoccal pneumonia), Staphylococcus aureus (staphylococcal pneumonia), Gram-negative bacterial pneumonia (caused by, e.
g.
,Klebsiella and Pseudomas spp.), Mycoplasma pneumoniae pneumonia, Hem~philus influenzae pneumonia, Legionella pneumophila (Legionnaires' disease), and Chlamydia psittaci (Psittacosis)), and viral pneumonia (e.g., influenza, chickenpox (varicella).
Additional diseases and disorders of the respiratory system include, but are not limited to bronchiolitis, polio (poliomyelitis), croup, respiratory syncytial viral infection, mumps, erythema infectiosum (fifth disease), roseola infantum, progressive rubella panencephalitis, german measles, and subacute sclerosing panencephalitis), fungal pneumonia (e.g., His~toplasmosis, Coccidioidomycosis, B,lastomycosis, fungal infections in people with severely suppressed immune systems (e.g., cryptococcosis, caused by CryptococcLCS neoformans; aspergillosis, caused by Aspergillus spp.;
candidiasis, caused by Candida; and mucormycosis)), Pneumocystis carinii (pneumocystis pneumonia), atypical pneumonias (e.g., Mycoplasma and Chlamydia spp.),, opportunistic infection pneumonia, nosocomial pneumonia, chemical pneumonitis, and aspiration pneumonia,. pleural disorders (e.g., pleurisy, pleural effusion, and pneumothorax (e.g., simple spontaneous .
pneumothorax, complicated spontaneous pneumothorax, tension pneumothorax)), obstructive airway diseases (e.g., asthma; chronic obstructive pulmonary disease (COPD), emphysema, chronic or acute bronchitis), occupational lung diseases (e.g., silicosis, black lung (coal workers' pneumoconiosis), asbestosis, berylliosis, occupational asthsma, byssinosis, and benign pneumoconioses), Infiltrative Lung Disease (e.g., ~
pulmonary fibrosis (e.g., fibrosing alveolitis, usual interstitial pneumonia), idiopathic pulmonary fibrosis, desquamative interstitial pneumonia, lymphoid interstitial pneumonia, histiocytosis X (e.g., Letterer-Siwe disease, Hand-Schiiller-Christian disease, eosinophilic granuloma), idiopathic pulmonary hemosiderosis, sarcoidosis and pulmonary alveolar proteinosis), Acute respiratory distress syndrome (also called, e.g., adult respiratory distress syndrome), edema, pulmonary embolism, bronchitis (e.g., viral, bacterial), bronchiectasis, atelectasis, lung abscess (caused by, e.g., Staphylococcus aureus or Legionella pneumophila), and cystic fibrosis.

Anti-Angiogenesis Activitx The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences predominate. Rastinejad et al., Cell 56:345-355 (1989). In those rare instances in which neovascularization occurs under normal physiological conditions, such as wound healing, organ regenei~ation, embryonic development,. and female reproductive processes, angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail.
Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases. A number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and metastases, arthritis, some types of eye disorders, and psoriasis. See, e.g., reviews by Moses et al., Biotech. 9:630-634 (1991);
Folkman et al., N. Engl. J. Med., 333:1757-1763 (1995); Auerbach et al., J.
Microvasc.
Res. 29:401-411 (1985); Folkman, Advances in Cancer Research, eds. Klein ..and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J.
Opthalmol.
94:715-743 (1982); and Folkman et al., Science 221:719-725 (1983). In a number of pathological conditions, the process of angiogenesis contributes to the disease state. For example, significant data have accumulated which suggest that the growth of solid,tumors is dependent on angiogenesis. Folkman and Klagsbrun, Science 235:442-447 (1987):
. The present invention provides for treatment of diseases or disorders associated with neovascularization by administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Malignant and metastatic conditions which can be treated with the polynucleotides and polypeptides, or agonists or antagonists of the invention include, but are not limited to, malignancies, solid tumors, and cancers described herein and otherwise known in the art (for a review of such disorders, see Fishman et al.,. Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).Thus, the present invention provides a method of treating an angiogenesis-related disease 'and/or disorder, comprising administering to an individual in need thereof a therapeutically effective amount of an albumin fusion protein of the ,invention and/or polynucleotides encoding an albumin fusion protein of the invention. For example, fusion proteins of the invention and/or polynucleotides encoding albumin -fusion proteins of .the invention may be utilized in a variety of additional methods in order to therapeutically treat a cancer or tumor. Cancers which may be treated with fusion proteins of the invention andlor polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to solid tumors, including prostate, lung, breast, ovarian, stomach, pancreas, _ larynx, esophagus,. testes, liver, parotid, biliary tract, colon, rectum, cervix, uterus, endometrium, kidney, bladder, thyroid cancer; primary tumors and metastases;
melanomas;
glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non- small cell lung cancer;
colorectal cancer; advanced malignancies; and blood bortl~tumors such as leukemias. For example, fusion proteins of the invention and/or polyriucleotides encoding albumin fusion proteins of the invention may be delivered topically, in order to treat cancers such as skin cancer, head and neck tumors, breast tumors, and Kaposi's sarcoma.
Within yet other aspects, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized to treat superficial forms of bladder cancer by, for example, intravesical administration. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be delivered directly into the tumor, or near the tumor site, via injection or a catheter. Of course, as the artisan of ordinary .skill will appreciate, the appropriate mode of administration will vary according to the cancer to be treated. Other modes of delivery are discussed herein.
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful. in treating other'disorders, besides cancers, which involve angiogenesis. These disorders include, but are not limited to:
benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; artheroscleric 'plaques; ocular angiogenic diseases, for example, diabetic 20- retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis;
delayed wound healing; endometriosis; vasculogenesis; granulations; hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma; vascular, adhesions; myocardial angiogenesis; coronary collaterals; cerebral collaterals; arteriovenous malformations; ischemic limb angiogenesis;
Osler-Webber~ Syndrome; plaque neovascularization; telangiectasia; hemophiliac joints;
angiofibroma; , fibromuscular dysplasia; wound granulation; Crohn's disease;
and ' atherosclerosis.
For example, within one aspect of the present invention methods are provided for treating hypertrophic scars and keloids, comprising the step of administering albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to a hypertrophic scar or keloid.
Within one embodiment of the present invention fusion proteins of the invention and/or poiynucleotides encoding albumin fusion proteins of the invention are directIy~
injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., burns) and is preferably initiated after the proliferative phase has had time to progress (approximately 14 days after the initial injury), but before hypertrophic scar or keloid development. As noted above, the present invention also provides methods for treating neovascular diseases of the eye, including for example, corneal neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration.
Moreover, Ocular disorders associated with neovascularization which can be treated with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to: neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of IO prerilaturity macular degeneration, corneal graft neovascularization, as well as other eye inflammatory diseases, ocular tumors and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman et al., Am. J. Ophthal.
X5:704-710 (1978) and Gartner et al., Surv. (7phthal. 22:291-312 (1978).
Thus within one aspect of the present invention methods are provided for treating neovascular diseases of the eye such as corneal neovascularization (including corneal graft neovascularization), comprising the step of administering to a patient a therapeutically effective amount of a compound (e.g., fusion, proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention) to the cornea, such that the formation of blood vessels is inhibited. Briefly, the cornea is a tissue which normally lacks blood vessels. In certain pathological conditions~however, capillaries may extend into the cornea from the pericorneal vascular plexus of the limbus. When the cornea becomes vascularized, it also becomes clouded, resulting in a decline in the patient's visual acuity.
Visual loss may become complete if the cornea completely opacitates. A wide variety of disorders can result in corneal neovascularization, including for example, corneal infections (e.g., trachoma, herpes simplex keratitis, leishmaniasis ,and onchocerciasis), immunological processes (e.g., graft rejection and Stevens-Johnson's syndrome), alkali burns, trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of wearing contact lenses.
Within particularly preferred embodiments of the invention, may be prepared for . topical administration in saline (combined with any of the preservatives and antimicrobial agents commonly used in ocular preparations), and administered in eyedrop form. The solution or suspension may be prepared in its pure form and administered several times .
daily. Alternatively, anti-angiogenic compositionsz prepared as'described above, may also be administered directly to the cornea. Within preferred embodiments, the anti-angiogenic composition is prepared with a muco-adhesive polymer which binds to~ cornea.
VWithin further embodiments, the' anti-angiogenic factors or .anti-angiogenic compositions may be utilized as an adjunct to conventional steroid therapy. Topical therapy may also be useful prophylactically in corneal lesions which are known to have a high probability of inducing an angiogenic response (such as chemical burns). In these instances the treatment, likely in combination with steroids, may be instituted immediately to help prevent subsequent complications.
Within other embodiments, the compounds. described above may be injected directly into the corneal'stroma by an ophthalmologist under microscopic guidance. The preferred site of injection may vary with the morphology of the individual lesion, but the goal of the administration would be to place the composition at the advancing front of the vasculature (i.e., interspersed between the blood vessels and the normal cornea). In most cases this would involve perilimbic corneal injection to "protect" the cornea from the advancing blood vessels. This method may also be utilized shortly after 'a corneal insult in order to prophylactically prevent corneal neoVascularization. In this situation the material could be injected in the,perilimbic cornea interspersed between the corneal lesion and its undesired potential limbic blood supply. Such methods may also be utilized in a similar fashion to prevent capillary invasion of .transplanted corneas. In a sustained-release form injections might only be required 2-3 times per year. A steroid could also be added to the injection solution to reduce inflammation resulting from the injection itself.
Within another aspect of the present invention, methods are provided for treating neovascular glaucoma, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the eye, such that the formation of blood vessels is inhibited. In one embodiment, the compound may be administered topically to the eye in order=to treat early forms of neovascular glaucoma.
Within other embodiments, the compound may be implanted by injection into the region of the anterior chamber angle. Within other embodiments, the compound may also be placed in any location such that the compound is continuously released into the aqueous humor. Within another aspect of the present invention, methods are provided for treating proliferative diabetic retinopathy, comprising the step of administering ~ to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the~eyes, such that the formation of blood vessels is inhibited.
Within particularly' preferred embodiments of the invention, proliferative diabetic retinopathy may be treated by injection into the aqueous humor or the vitreous, in order to increase the local concentration of the polynucleotide, polypeptide, antagonist and/or agonist in the retina. Preferably, this treatment should be initiated prior to the acquisition of severe ' disease requiring photocoagulation. .
Within another aspect of the present invention, methods are provided for treating 224 ~ ' retrolental fibroplasia, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of .the invention to the eye, such that the formation of - blood vessels is inhibited. The compound may be administered topically, via intravitreous injection and/or via intraocular implants.
Additionally, disorders which can be treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion 'proteins of the invention include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations; hemophilic joints,' hypertrophic scars, nonunion fractures, Osler-Weber syndrome, pyogenic granuloma, scleroderma, trachoma;
and vascular adhesions.
Moreover, disorders andlor states, which can be treated, prevented, diagnosed, and/or prognosed with the the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention of the invention include, I5 but are not limited to, solid tumors, blood born tumors such as ~leukemias, tumor metastasis, Kaposi's sarcoma, benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic diseases for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascuIar glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, and uvietis, delayed wound healing, endometriosis, vascluogenesis, granulations, hypertrophic scars (keloids), nonunion fractures, scleroderma, trachoma, vascular adhesions, myocardial angiogenesis, coronary collaterals,' cerebral collaterals, arteriovenous malformations, ischemic limb angiogenesis, Osler- ' ' Webber Syndrome, plaque neovascularization, telangiectasia, hemophiliac joints, angiofibroma fibromuscular dysplasia, wound granulation, Crohn's disease, atherosclerosis, birth control agent by preventing vascularization required for embryov implantation controlling menstruation, diseases that have angiogenesis as a pathologic consequence such as cat scratch disease (Rochele minalia quintosa), ulcers (Helicobacter pylori), Bartonellosis and baci~llary angiomatosis. ~ ' ' In one aspect of the birth control method, an amount of the compound sufficient to block embryo implantation is administered before or after intercourse and fertili-zation have occurred, thus providing an effective method of birth control, possibly a "morning after"
method. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be ,used in controlling menstruation or administered as either a peritoneal lavage fluid or for peritoneal implantation in the treatment of endometriosis.
Albumin fusion proteins of the invention and/or polynucleotides encoding, albumin fusion proteins of the invention may be incorporated into surgical sutures in order_to prevent ~_stitch granulomas.
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized in a wide variety of surgical procedures.
For example, within one aspect of the present invention a compositions (in the form of, for example, a spray or film) may be utilized to coat or spray an area prior to removal of a tumor, in order to isolate normal surrounding tissues from malignant tissue, and/or to prevent the spread of disease to surrounding tissues. Within other aspects of the present invention, compositions (e.g., in the form of a spray) may be delivered via endoscopic procedures in order to coat tumors, or inhibit angiogenesis in a desired locale. Within yet other aspects of the present invention, surgical meshes which have been coated with anti-angiogenic compositions of the present invention may be utilized in any procedure wherein a surgical mesh might be utilized. . For example, within one embodiment of the invention a surgical mesh laden with an anti-angiogenic composition may be utilized during abdominal cancer resection surgery (e.g., subsequent to colon resection) in order to provide support to the structure, and to release an amount of the anti-angiogenic factor.
Within further aspects of the . present invention, methods are provided for ,treating tumor excision sites, comprising administering albumin fusion proteins of the invention and/or polynucl.eotides encoding albumin fusion proteins 'of the invention to the resection margins of a tumor subsequent to~excision, such that the local recurrence of cancer and the formation of new blood vessels at the site is inhibited. Within one embodiment of the invention, the anti-angiogenic compound is administered directly to the tumor excision site (e.g., applied by swabbing, brushing or otherwise coating the resection margins of the tumor with the anti-angiogenic compound). Alternatively, the anti-angiogenic compounds may be incorporated into known surgical pastes prior to administration. Within particularly preferred embodiments of the invention, the anti-angiogenic 'compounds. are applied after hepatic resections for malignancy, and after neurosurgical operations.
Within one aspect of the present invention,, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be administered to the resection margin of a wide variety of tumors, including for example, breast, colon, brain and hepatic tumors. For example, within one embodiment of the invention, anti-angiogenic compounds may be administered to the site of a. neurological tumor subsequent to excision, such that the formation of new blood vessels at the site are inhibited.
The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be administered along with other anti-angiogenic factors. Representative examples of other anti-angiogenic factors include: Anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel, Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, Plasminogen Activator Inhibitor-l, Plasminogen Activator Inhibitor-2, and various forms of the lighter "d group"
transition metals.
Lighter "d group" transition metals include, for example, vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition metal species may form transition metal complexes. Suitable complexes of the above-mentioned transition metal species include oxo transition' metal complexes.
Representative examples of vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes. Suitable vanadate complexes include ~ metavanadate and orthovanadate complexes such as, for example, ammonium metavanadate, sodi,urri metavanadate, and sodium orthovanadate. Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates.
Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes. Suitable tungstate complexes include ammonium tungsfate, calcium tungstate, sodium tungstate dehydrate, and tungstic acid. Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) ~ oxide.- Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates. Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum (VI) oxide, and molybdic acid. Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate. Other suitable tungsten and molybdenum complexes include. hydroxo derivatives derived from, for example, glycerol, tartaric acid, and ugars.
A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include' platelet factor 4;
protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells) (Murata et al., Cancer Res. 51:22-26, 1991); Sulphated Polysaccharide Peptidoglycan Complex (SP- PG) (the~funetion of this compound may be enhanced by the presence of steroids such as, estrogen, and tamoxifen citrate); Staurosporine; modulators of. matrix metabolism, including for example, proline' analogs,. cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-' 2(3H)-oxazolone; Methotrexate; Mitoxantrone; ~ Heparin; Interferons; 2 Macroglobulin serum; ChIMP-3 (Pavloff et al., 1J. Bio. Chem. 267:17321-17326, (1992));
Chymostatin (Tomkinson et al., Biochem J. 286:475-480, (1992)); Cyclodextrin Tetradecasulfate;
Eponemycin; Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, 1990); Gold Sodium Thiomalate ("GST"; Matsubara and Ziff, J. Clin. Invest. 79:1440-1446, (1987));
anticollagenase-serum; alpha2-antiplasmin (Holmes et al., J. Biol. Chem.
262(4):1659-1664, (1987)); Bisantrene (National Cancer InstiWte); Lobenzarit disodium (N-(2)-' carboxyphenyl-4- chloroanthronilic acid disodium or "CCA"; Takeuchi et al., Agents Actions 36:312-316, (1992)); Thalidomide; Angostatic steroid; AGM-1470;
carboxynaminolmidazole; and metalloproteinase inhibitors such as BB94.
Diseases at the Cellular Level Diseases associated with increased cell survival or the inhibition of apoptosis that could be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, ' stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's ~thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection. ' In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to inhibit growth, progression, and/or metasis'of cancers, i.n particular those listed above.
Additional diseases or conditions associated with increased cell survival that could be treated or detected by fusion proteins of the invention and/or polynucleotides encoding, albumin fusion proteins of the invention include, but are not limited to;
progression, andlor metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute -myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic ' (granulocytic) leukemia and chronic lymphocytic ~ leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple. myeloma, Waldenstrom's macroglobulinemia, heavy .
chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma;
chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, . bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small' cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
Diseases associated with increased apoptosis that could be treated, prevented, diagnosed, and/or prognesed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, AIDS;
neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis,. Behcet's disease, Crohn's disease, .
polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury (such as that caused . by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
Wound Healing and Epithelial Cell Proliferation In accordance with- yet a further aspect of the present invention, there is piovided a process for utilizing fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, for therapeutic purposes, for example, to stimulate epithelial cell proliferation and basal keratinocytes for the purpose of wound healing, and to stimulate hair follicle production and healing of dermal wounds. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be clinically useful in stimulating wound healing including surgical wounds, excisional wounds, deep wounds involving damage of the dermis and epidermis, eye tissue wounds, ' 35 dental tissue wounds, oral cavity wounds, diabetic ulcers, dermal ulcers, cubitus ulcers, arterial ulcers,-venous stasis ulcers, burns resulting from heat exposure or chemicals, and other abnormal wound healing conditions such as uremia, malnutrition, vitamin deficiencies DEMANDE OU BREVET VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

~~ TTENANT LES PAGES 214 A 229 NOTE : Pour les tomes additionels, veuillez contacter 1e 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 NOM DU FICHIER / FILE NAME
NOTE POUR LE TOME / VOLUME NOTE:

Claims (36)

What is claimed:
1. An albumin fusion protein comprising a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:18.
2. An albumin fusion protein comprising a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:18, wherein said fragment or variant has albumin activity.
3. The albumin fusion protein of claim 2, 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.
4. The albumin fusion protein of claim 2, wherein the fragment or variant comprises the amino acid sequence of amino acids 1-387 of SEQ ID NO:18.
5. An albumin fusion protein comprising a fragment or variant of a Therapeutic protein:X, and albumin comprising the amino acid sequence of SEQ
ID
NO:18, wherein said fragment or variant has a biological activity of the Therapeutic protein:X.
6. The albumin fusion protein of any one of claims 1-5, 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. .
7. The albumin fusion protein of any one of claims 1-5, 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..
8. The albumin fusion protein of any one of claims 1-5, 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-terminus of the fragment or variant of albumin.
9. The albumin fusion protein of any one of claims 1-5, 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.
10. The albumin fusion protein of any one of claims 1-8, 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.
11. The albumin fusion protein of any one of claims 1-8, wherein the albumin fission protein has the following formula:
R1-L-R2; R2-L-R1; or R1-L-R2-L-R1, 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:18 or fragment or variant of albumin.
12. The albumin fusion protein of any one of claims 1-11, wherein the shelf-life of the albumin fusion protein is greater than the shelf-life of the Therapeutic protein:X in an unfused state.
13. The albumin fusion protein of any one of claims 1-11, wherein the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin, or fragment or variant thereof, is greater than the in vitro biological activity of the Therapeutic protein:X, or a fragment or variant thereof, in an unfused state.
14. The albumin fusion protein of any one of claims 1-11, wherein the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin, or fragment or variant thereof, is greater than the in vivo biological activity of the Therapeutic protein:X , or a fragment or variant thereof, in an unfused state.
15. An albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof, inserted into an albumin comprising the amino acid sequence of SEQ ID
NO:18 or fragment or variant thereof.
16. An albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof, inserted into an albumin comprising an amino acid sequence selected from the group consisting of:
(a) amino acids 54 to 61 of SEQ ID NO:18;
(b) amino acids 76 to 89 of SEQ ID NO:18;
(c) amino acids 92 to 100 of SEQ ID NO:18;
(d) amino acids 170 to 176 of SEQ ID NO:18;
(e) amino acids 247 to 252 of SEQ ID NO:18;
(f) amino acids 266 to 277 of SEQ ID NO:18; .
(g) amino acids 280 to 288 of SEQ ID NO:18;
(h) amino acids 362 to 368 of SEQ ID NO:18;
(i) amino acids 439 to 447 of SEQ ID NO:18;
(j) amino acids 462 to 475 of SEQ ID NO:18;
(k) amino acids 478 to 486 of SEQ ID NO:18; and (l) amino acids 560 to 566 of SEQ ID NO:18.
17. The albumin fusion protein of claims 15 or 16, wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, as compared to the shelf-life of the Therapeutic protein:X , or a fragment or variant thereof, in an unfused state.
18. The albumin fusion protein of claims 15 or 16, wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin as compared to the in vitro biological activity of the Therapeutic protein:X , or a fragment or variant thereof, in an unfused state.
19. The albumin fusion protein of claims 15 or 16 wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin compared to the in vivo biological activity of the Therapeutic protein:X , or a fragment or variant thereof, in an unfused state.
20. The albumin fusion protein of any one of claims 1-19, which is non-glycosylated.
21. The albumin fusion protein of any one of claims 1-19, which is expressed in yeast.
22. The albumin fusion protein of claim 21, wherein the yeast is glycosylation deficient.
23. The albumin fusion protein of claim 21 wherein the yeast is glycosylation and protease deficient.
24. The albumin fusion protein of any one of claims 1-19, which is expressed by a mammalian cell.
25. The albumin fusion protein of any one of claims 1-19, wherein the albumin fusion protein is expressed by a mammalian cell in culture.
26. The albumin fusion protein of any one of claims 1-19, wherein the albumin fusion protein further comprises a secretion leader sequence.
27. A composition comprising the albumin fusion protein of any one of claims 1-26 and a pharmaceutically acceptable carrier.
28. A kit comprising the composition of claim 27.
29. A method of treating a disease or disorder in a patient, comprising the step of administering the albumin fusion protein of any one of claims 1-26.
30. The method of claim 29, wherein the disease or disorder comprises indication:Y.
31. A method of treating a patient with a disease or disorder that is modulated by Therapeutic protein:X, or fragment or variant thereof, comprising the step of administering an effective amount of the albumin fusion protein of any one of claims 1-26.
32. The method of claim 31, wherein the disease or disorder is indication:Y.
33. A method of extending the shelf-life of Therapeutic protein:X comprising the step of fusing the Therapeutic protein:X, or fragment or variant thereof, to albumin or a 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 a fragment or variant thereof, in an unfused state.
34. A nucleic acid molecule comprising a polynucleotide sequence encoding the albumin fusion protein of any one of claims 1-26.
35. A vector comprising the nucleic acid molecule of claim 34.
36. A host cell comprising the nucleic acid molecule of claim 35.
CA002405550A 2000-04-12 2001-04-12 Albumin fusion proteins Abandoned CA2405550A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US22935800P 2000-04-12 2000-04-12
US19938400P 2000-04-25 2000-04-25
US25693100P 2000-12-21 2000-12-21
PCT/US2001/011850 WO2001079442A2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins

Publications (1)

Publication Number Publication Date
CA2405550A1 true CA2405550A1 (en) 2001-10-25

Family

ID=27394014

Family Applications (8)

Application Number Title Priority Date Filing Date
CA2747325A Abandoned CA2747325A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405701A Withdrawn CA2405701A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405525A Abandoned CA2405525A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405709A Abandoned CA2405709A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405550A Abandoned CA2405550A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405563A Abandoned CA2405563A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA2405557A Expired - Lifetime CA2405557C (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405912A Abandoned CA2405912A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins

Family Applications Before (4)

Application Number Title Priority Date Filing Date
CA2747325A Abandoned CA2747325A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405701A Withdrawn CA2405701A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405525A Abandoned CA2405525A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405709A Abandoned CA2405709A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins

Family Applications After (3)

Application Number Title Priority Date Filing Date
CA002405563A Abandoned CA2405563A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA2405557A Expired - Lifetime CA2405557C (en) 2000-04-12 2001-04-12 Albumin fusion proteins
CA002405912A Abandoned CA2405912A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins

Country Status (10)

Country Link
US (23) US6926898B2 (en)
EP (21) EP2275557A1 (en)
JP (9) JP2003530838A (en)
AU (7) AU2001261024A1 (en)
BE (1) BE2016C059I2 (en)
CA (8) CA2747325A1 (en)
DK (2) DK2216409T3 (en)
ES (2) ES2484966T3 (en)
FR (1) FR16C0043I2 (en)
WO (7) WO2001079442A2 (en)

Families Citing this family (639)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2686899B1 (en) * 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US6057287A (en) 1994-01-11 2000-05-02 Dyax Corp. Kallikrein-binding "Kunitz domain" proteins and analogues thereof
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
WO2002018435A1 (en) * 2000-08-28 2002-03-07 Human Genome Sciences, Inc. 18 human secreted proteins
WO2002016576A1 (en) * 2000-08-18 2002-02-28 Human Genome Sciences, Inc. 11 human secreted proteins
US20020137890A1 (en) 1997-03-31 2002-09-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP0892047A3 (en) * 1997-07-09 2000-03-08 Hoechst Marion Roussel Deutschland GmbH Human and murine semaphorin L
US20030166108A1 (en) * 1997-09-18 2003-09-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
CA2330527A1 (en) * 1998-06-15 1999-12-23 Genzyme Transgenics Corporation Erythropoietin analog-human serum albumin fusion
US20050181482A1 (en) * 2004-02-12 2005-08-18 Meade Harry M. Method for the production of an erythropoietin analog-human IgG fusion proteins in transgenic mammal milk
US20040199099A1 (en) * 1998-07-10 2004-10-07 Matson James R Hemofiltration systems, methods and devices used to treat inflammatory mediator related disease
US6818611B1 (en) 1998-10-13 2004-11-16 University Of Georgia Research Foundation, Inc. Stabilized bioactive peptides and methods of identification, synthesis and use
US20030190740A1 (en) 1998-10-13 2003-10-09 The University Of Georgia Research Foundation, Inc Stabilized bioactive peptides and methods of identification, synthesis, and use
CA2721199A1 (en) * 1999-05-05 2000-11-16 Phylogica Limited Isolating biological modulators from biodiverse gene fragment libraries
US6946129B1 (en) 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
EP1224285A4 (en) * 1999-10-29 2004-12-08 Human Genome Sciences Inc 27 human secreted proteins
CA2388019A1 (en) * 1999-11-05 2001-05-17 Human Genome Sciences, Inc. 24 human secreted proteins
JP2003520030A (en) * 1999-11-05 2003-07-02 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 28 human secreted proteins
EP1235844A4 (en) * 1999-11-12 2005-01-19 Human Genome Sciences Inc 28 human secreted proteins
CA2392477A1 (en) 1999-11-30 2001-06-07 Mayo Foundation For Medical Education And Research B7-h1, a novel immunoregulatory molecule
ATE337403T1 (en) 1999-12-24 2006-09-15 Genentech Inc METHOD AND COMPOUNDS FOR EXTENSING THE HALF-LIFE TIMES IN THE EXCRETION OF BIOACTIVE COMPOUNDS
US7534417B2 (en) 2000-02-24 2009-05-19 Agensys, Inc. 103P2D6: tissue specific protein highly expressed in various cancers
US7291122B2 (en) * 2000-03-24 2007-11-06 Immunocept, L.L.C. Hemofiltration methods for treatment of diseases in a mammal
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20050100991A1 (en) * 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
CA2747325A1 (en) * 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US7030219B2 (en) * 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
US20030143654A1 (en) * 2000-05-12 2003-07-31 Matthias Grell F-box containing protein
US6787040B2 (en) * 2000-05-16 2004-09-07 Immunocept, L.L.C. Method and system for colloid exchange therapy
WO2001090179A2 (en) * 2000-05-23 2001-11-29 Lexicon Genetics Incorporated Novel human thrombospondin-like proteins and polynucleotides encoding the same
AU2001275285A1 (en) 2000-06-06 2001-12-17 Bristol-Myers Squibb Company B7-related nucleic acids and polypeptides and their uses for immunomodulation
EP1283886A2 (en) * 2000-06-08 2003-02-19 Incyte Genomics, Inc. Intracellular signaling proteins
MXPA02012717A (en) * 2000-06-21 2003-09-22 Amgen Inc Secreted epithelial colon stromal-1 polypeptides, nucleic acids encoding the same and uses thereof.
CA2418676A1 (en) * 2000-08-18 2002-02-28 Human Genome Sciences, Inc. 21 human secreted proteins
CA2419306A1 (en) * 2000-08-18 2002-02-28 Human Genome Sciences, Inc. 23 human secreted proteins
CA2420192A1 (en) * 2000-09-20 2002-03-28 Human Genome Sciences, Inc. 21 human secreted proteins
AU2001227920A1 (en) * 2000-09-29 2002-04-15 Human Genome Sciences, Inc. 24 human secreted proteins
CA2425827A1 (en) * 2000-10-12 2002-04-18 Hyseq, Inc. Novel nucleic acids and polypeptides
US6949371B2 (en) 2000-10-20 2005-09-27 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6531297B2 (en) * 2000-10-20 2003-03-11 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
FR2815964A1 (en) * 2000-10-30 2002-05-03 Inst Nat Sante Rech Med RENIN AND / OR PRORENIN RECEPTOR PROTEIN, NUCLEIC ACID ENCODING THE RECEPTOR AND THEIR APPLICATIONS
EP1395657B1 (en) 2000-12-05 2007-04-18 Wisconsin Alumni Research Foundation Receptor for bacillus anthracis toxin
BR0116024A (en) 2000-12-07 2005-12-13 Lilly Co Eli Heterologous Fusion Protein and Use thereof
US20030148920A1 (en) * 2000-12-27 2003-08-07 Steven Rosen Sulfatases and methods of use thereof
JP4344519B2 (en) * 2000-12-28 2009-10-14 旭化成ファーマ株式会社 NF-κB activating gene
CA2433469A1 (en) * 2001-02-23 2002-09-06 Human Genome Sciences, Inc. 83 human secreted proteins
WO2002072138A1 (en) * 2001-03-08 2002-09-19 Hyseq, Inc. Methods and materials relating to fibulin-like polypeptides and polynucleotides
WO2002083921A2 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
US7507413B2 (en) * 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
US20060084794A1 (en) * 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
US7164007B2 (en) 2001-06-20 2007-01-16 Genentech, Inc. Anti-PR020044 antibodies
US20040234537A1 (en) * 2001-07-06 2004-11-25 Lydie Bougueleret Carcinoma-related peptides
US8129504B2 (en) 2001-08-30 2012-03-06 Biorexis Technology, Inc. Oral delivery of modified transferrin fusion proteins
US7176278B2 (en) 2001-08-30 2007-02-13 Biorexis Technology, Inc. Modified transferrin fusion proteins
AU2002332041A1 (en) * 2001-10-05 2003-04-22 Human Genome Sciences, Inc. Albumin fusion proteins
DE10251673A1 (en) 2001-11-09 2003-07-10 Hoffmann La Roche Alström syndrome gene, gene variants, encoded protein and method for diagnosing Alström syndrome
KR100406760B1 (en) * 2001-11-16 2003-11-21 신코엠 주식회사 Semiconductor memory device
EP1458756B1 (en) 2001-12-17 2009-01-28 Laboratoires Serono SA Chemokine mutants acting as chemokine antagonists
US20040082761A1 (en) * 2001-12-18 2004-04-29 Duggan Brendan M. Cell adhesion proteins
EP1463751B1 (en) 2001-12-21 2013-05-22 Human Genome Sciences, Inc. Albumin fusion proteins
WO2005003296A2 (en) * 2003-01-22 2005-01-13 Human Genome Sciences, Inc. Albumin fusion proteins
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
US20080194481A1 (en) * 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
US20060253913A1 (en) * 2001-12-21 2006-11-09 Yue-Jin Huang Production of hSA-linked butyrylcholinesterases in transgenic mammals
EP1463752A4 (en) * 2001-12-21 2005-07-13 Human Genome Sciences Inc Albumin fusion proteins
US7081446B2 (en) * 2002-01-31 2006-07-25 The Trustees Of Columbia University In The City Of New York Long-acting follicle stimulating hormone analogues and uses thereof
ZA200406124B (en) * 2002-02-07 2008-09-25 Delta Biotechnology Ltd Albumin-fused kunitz domain peptides
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
DE10205520A1 (en) * 2002-02-08 2003-08-14 Aventis Behring Gmbh Inhibitory, monoclonal antibody against the clotting factor VII activating protease
KR20040096592A (en) * 2002-02-21 2004-11-16 와이어쓰 Follistatin domain containing proteins
US20050169839A1 (en) * 2002-03-05 2005-08-04 Fong Tung M. Biomarker for efficacy of appetite suppressant drugs
US20030191056A1 (en) * 2002-04-04 2003-10-09 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
US20070015230A1 (en) * 2002-04-15 2007-01-18 Hammond David J Identification and characterization of analytes from whole blood
AU2003225008A1 (en) * 2002-04-15 2003-11-03 American National Red Cross Plasma protein-binding ligands
AU2003234136A1 (en) * 2002-04-18 2003-11-03 The General Hospital Corporation Drg11-responsive (dragon) gene family
US7141381B2 (en) 2002-04-25 2006-11-28 Bristol-Myers Squibb Company Human leucine-rich repeat-containing proteins specifically expressed in the nervous system
GB0210464D0 (en) 2002-05-08 2002-06-12 Svanborg Catharina Therapeutic treatment
US7153829B2 (en) 2002-06-07 2006-12-26 Dyax Corp. Kallikrein-inhibitor therapies
JP2005534647A (en) 2002-06-07 2005-11-17 ダイアックス、コープ Prevention and reduction of blood loss
EP2377549A1 (en) * 2002-06-07 2011-10-19 ZymoGenetics, Inc. Use of IL-21 for treating viral infections
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US8029803B2 (en) 2002-06-20 2011-10-04 Paladin Labs, Inc. Chimeric antigens for eliciting an immune response
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
WO2004005520A1 (en) * 2002-07-03 2004-01-15 Nexgen Biotechnologies, Inc. Method for preparing fusion polypeptide comprising epidermal growth factor and human serum albumin in plants
CA2489588A1 (en) * 2002-07-08 2004-01-15 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
AU2003281626A1 (en) * 2002-07-22 2004-02-09 Masayoshi Harigai Novel gene associated with rheumatoid arthritis
GB0217033D0 (en) 2002-07-23 2002-08-28 Delta Biotechnology Ltd Gene and polypeptide sequences
EP1529108A2 (en) * 2002-08-07 2005-05-11 ZLB Behring GmbH Albumin-fused ciliary neurotrophic factor
AU2003255276A1 (en) * 2002-08-13 2004-02-25 Arbios Systems, Inc. Selective plasma exchange therapy
EP2386310B1 (en) 2002-08-28 2018-11-07 Dyax Corp. Methods for preserving organs and tissues
EP1545611B1 (en) * 2002-09-06 2016-11-09 Alexion Pharmaceuticals, Inc. Method of treatment of asthma using antibodies to complement component c5
US20050271660A1 (en) 2002-09-06 2005-12-08 Alexion Pharmaceuticals, Inc. Nebulization of monoclonal antibodies for treating pulmonary diseases
US9415102B2 (en) 2002-09-06 2016-08-16 Alexion Pharmaceuticals, Inc. High concentration formulations of anti-C5 antibodies
WO2004027064A2 (en) * 2002-09-18 2004-04-01 Centre Hospitalier De L'universite De Montreal (Chum) Ghrh analogues
US7432351B1 (en) 2002-10-04 2008-10-07 Mayo Foundation For Medical Education And Research B7-H1 variants
DE10254601A1 (en) 2002-11-22 2004-06-03 Ganymed Pharmaceuticals Ag Gene products differentially expressed in tumors and their use
EP1572739B1 (en) * 2002-12-20 2008-07-16 Geneos OY Asthma susceptibility locus
WO2004058817A1 (en) * 2002-12-26 2004-07-15 Takeda Pharmaceutical Company Limited Novel proteins and use thereof
MXPA05008533A (en) * 2003-02-11 2005-10-20 Transkaryotic Therapies Inc Diagnosis and treatment of multiple sulfatase deficiency and other using a formylglycine generating enzyme (fge).
EP1605961A4 (en) * 2003-03-12 2009-11-11 Vasgene Therapeutics Inc Polypeptide compounds for inhibiting angiogenesis and tumor growth
US7381410B2 (en) * 2003-03-12 2008-06-03 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
WO2004082640A2 (en) * 2003-03-19 2004-09-30 New Century Pharmaceuticals, Inc. Human serum albumin conjugates with therapeutic compounds
JP2007527206A (en) * 2003-04-04 2007-09-27 ユニバーシティ オブ ローザンヌ Peptabody for cancer treatment
KR20110094361A (en) 2003-04-11 2011-08-23 메디뮨 엘엘씨 Recombinant il-9 antibodies and uses thereof
WO2004094589A2 (en) * 2003-04-18 2004-11-04 Incyte Corporation Secreted proteins
US20050079546A1 (en) * 2003-05-01 2005-04-14 Dasa Lipovsek Serum albumin scaffold-based proteins and uses thereof
US6987270B2 (en) 2003-05-07 2006-01-17 General Electric Company Method to account for event losses due to positron range in positron emission tomography and assay of positron-emitting isotopes
WO2005001093A1 (en) * 2003-06-30 2005-01-06 Nihon University Protein capable of deposition onto extracellular matrix
CA2532250A1 (en) 2003-07-15 2005-02-03 Barros Research Institute Compositions and methods for immunotherapy of cancer and infectious diseases
US8007805B2 (en) * 2003-08-08 2011-08-30 Paladin Labs, Inc. Chimeric antigens for breaking host tolerance to foreign antigens
GB0320877D0 (en) * 2003-09-05 2003-10-08 Celltech R&D Ltd A protein involved in carcinoma
PL1682178T3 (en) 2003-11-04 2010-12-31 Novartis Vaccines & Diagnostics Inc Methods of therapy for cancers expressing the cd40 antigen
US20080292581A1 (en) * 2003-12-03 2008-11-27 Delta Biotechnology Limited Interleukin-11 Fusion Proteins
CN100379762C (en) * 2003-12-08 2008-04-09 中国人民解放军军事医学科学院生物工程研究所 Interfusion protein between human serum albumin and interleukin, and encoding genes
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US7371381B2 (en) 2003-12-12 2008-05-13 Amgen Inc. Anti-galanin antibodies and uses thereof
GB0329681D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Gene expression technique
GB0329722D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Modified plasmid and use thereof
EP1712619A4 (en) * 2003-12-24 2008-01-02 Takeda Pharmaceutical Preventive/remedy for cancer
PL1729795T3 (en) * 2004-02-09 2016-08-31 Human Genome Sciences Inc Albumin fusion proteins
SE0400489D0 (en) * 2004-02-27 2004-02-27 Biovitrum Ab Therapeutic proteins
WO2005085472A2 (en) * 2004-03-03 2005-09-15 Evotec Neurosciences Gmbh Diagnostic and therapeutic use of mal2 gene and protein for neurodegenerative diseases
US20050201959A1 (en) * 2004-03-11 2005-09-15 Vvii Newco 2003, Inc. Methods and compositions for altering skin coloration
US20080044439A1 (en) * 2004-03-11 2008-02-21 David Nathaniel E Compositions and Methods for Preventing and Treating Skin and Hair Conditions
ATE492564T1 (en) 2004-03-12 2011-01-15 Vasgene Therapeutics Inc EPHB4-BINDING ANTIBODIES FOR INHIBITING ANGIOGENESIS AND TUMOR GROWTH
EP1734991A4 (en) 2004-04-14 2012-10-24 Avirid Inc Compositions with modified nucleases targeted to viral nucleic acids and methods of use for prevention and treatment of viral diseases
CA2564031A1 (en) * 2004-04-23 2005-11-03 Conjuchem Biotechnologies Inc. Method for the purification of albumin conjugates
KR100599454B1 (en) * 2004-04-27 2006-07-12 재단법인서울대학교산학협력재단 3 Novel use of AIM3 acting as a tumor suppressor
DE102004024617A1 (en) 2004-05-18 2005-12-29 Ganymed Pharmaceuticals Ag Differentially expressed in tumors gene products and their use
CA2568976A1 (en) * 2004-06-03 2005-12-22 Ciphergen Biosystems, Inc. Biomarkers for peripheral artery disease
US7906140B2 (en) 2004-06-17 2011-03-15 Virun, Inc. Compositions for mucosal delivery of agents
JP2008506703A (en) 2004-07-14 2008-03-06 ユニバーシティ オブ ユタ リサーチ ファウンデーション Netrin-related compounds and uses
US20060068425A1 (en) * 2004-08-13 2006-03-30 Millennium Pharmaceuticals, Inc. Genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of prostate cancer
WO2006034292A2 (en) 2004-09-21 2006-03-30 Medimmune, Inc. Antibodies against and methods for producing vaccines for respiratory syncytial virus
AU2005286662B2 (en) * 2004-09-23 2011-10-06 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
US7235530B2 (en) 2004-09-27 2007-06-26 Dyax Corporation Kallikrein inhibitors and anti-thrombolytic agents and uses thereof
US20060078541A1 (en) * 2004-10-01 2006-04-13 Giles Brian C Method and formula for stem cells' stimulation, targeting release, trafficking and homing
MX2007004176A (en) * 2004-10-06 2007-06-15 Mayo Foundation B7-h1 and methods of diagnosis, prognosis, and treatment of cancer.
WO2006042197A2 (en) * 2004-10-11 2006-04-20 The Board Of Trustees Of The Leland Standford Junior University Use of del-1 in hair, bone and cartilage regeneration
US8071716B2 (en) 2004-10-25 2011-12-06 Immune System Key Ltd. Thymus-specific protein
KR100583350B1 (en) * 2004-11-03 2006-06-05 (주)넥스젠 -1 Method for Producing Epidermal Growth Factor Using Fusion Proteins Comprising Fas-1 Domain
DE602005009856D1 (en) * 2004-11-09 2008-10-30 Ares Trading Sa PROCESS FOR CLEANING FSH
JP5631533B2 (en) 2004-12-23 2014-11-26 ノボザイムス バイオファーマ デーコー アクティーゼルスカブ Gene expression technology
JP5855326B2 (en) 2005-01-06 2016-02-09 ノヴォ ノルディスク アー/エス Anti-KIR combination therapy and method
US20060178301A1 (en) * 2005-02-04 2006-08-10 Mathias Jurs Albumin-fused ciliary neurotrophic factor
AU2006214121B9 (en) 2005-02-15 2013-02-14 Duke University Anti-CD19 antibodies and uses in oncology
US20110230407A1 (en) * 2005-03-14 2011-09-22 Alexander Yuzhakov Hepatocyte growth factor pathway activators in demyelinating diseases and central nervous system trauma
WO2006107611A2 (en) * 2005-03-23 2006-10-12 Wyeth Detection of an immune response to gdf-8 modulating agents
BRPI0609449A2 (en) * 2005-03-23 2010-04-06 Wyeth Corp gdf-8 modulation agent detection
EP2360181B1 (en) 2005-04-18 2013-09-18 Novo Nordisk A/S IL-21 variants
US7833979B2 (en) * 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
JP5129122B2 (en) 2005-04-26 2013-01-23 トリオン ファーマ ゲーエムベーハー Combination of antibodies and glucocorticoids for cancer treatment
US9585932B2 (en) 2005-04-29 2017-03-07 Peter C. Dowling Use of EPO-derived peptide fragments for the treatment of neurodegenerative disorders
US9345745B2 (en) 2005-04-29 2016-05-24 Bo Wang Methods for treating inflammatory disorders and traumatic brain injury using stabilized non-hematopoietic EPO short peptides
WO2007052154A2 (en) * 2005-04-29 2007-05-10 University Of Medicine And Dentistry Of New Jersey Erythropoietin-derived short peptide and its mimics as immuno/inflammatory modulators
CA2607281C (en) 2005-05-05 2023-10-03 Duke University Anti-cd19 antibody therapy for autoimmune disease
US20070053903A1 (en) * 2005-05-12 2007-03-08 Zeren Gao Methods of using pHHLA2 to co-stimulate T-cells
BRPI0611766A2 (en) 2005-06-08 2011-12-20 Dana Farber Cancer Inst Inc methods and compositions for the treatment of persistent infections and cancer by inhibition of the programmed cell death pathway
WO2007014167A2 (en) * 2005-07-22 2007-02-01 Five Prime Therapeutics, Inc. Compositions for and methods of treating epithelial diseases with growth factors
US8323666B2 (en) * 2005-08-01 2012-12-04 Allergan, Inc. Botulinum toxin compositions
CA2618681C (en) 2005-08-10 2015-10-27 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
PT1917276T (en) * 2005-08-26 2018-06-11 Ares Trading Sa Process for the preparation of glycosylated interferon beta
US7713715B2 (en) * 2005-09-06 2010-05-11 University Of Tennessee Research Foundation Method for diagnosing infections
US7846445B2 (en) * 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
CA2622441A1 (en) * 2005-09-27 2007-04-05 Amunix, Inc. Proteinaceous pharmaceuticals and uses thereof
US20090099031A1 (en) * 2005-09-27 2009-04-16 Stemmer Willem P Genetic package and uses thereof
JP2009509564A (en) * 2005-10-03 2009-03-12 アストラゼネカ・アクチエボラーグ Fusion proteins with regulated plasma half-life
US8168592B2 (en) * 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
EP1790664A1 (en) 2005-11-24 2007-05-30 Ganymed Pharmaceuticals AG Monoclonal antibodies against claudin-18 for treatment of cancer
US20090304687A1 (en) * 2005-12-09 2009-12-10 Seattle Genetics , Inc. Methods of using cd40 binding agents
US8604175B2 (en) * 2005-12-09 2013-12-10 Ares Trading S.A. Method for purifying FSH or a FSH mutant
US8178495B2 (en) 2008-06-27 2012-05-15 Duke University Therapeutic agents comprising a GLP-1 receptor agonist and elastin-like peptide
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
CN101384623B (en) * 2005-12-22 2013-07-24 常山凯捷健生物药物研发(河北)有限公司 Process for the production of preformed conjugates of albumin and a therapeutic agent
US20090215084A1 (en) * 2006-01-05 2009-08-27 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
US20100015642A1 (en) * 2006-01-05 2010-01-21 Kwon Eugene D B7-h1 and survivin in cancer
CA2637600A1 (en) 2006-01-17 2007-07-26 Health Research, Inc. Heteroduplex tracking assay
EP1816201A1 (en) * 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
WO2007097961A1 (en) * 2006-02-16 2007-08-30 Massachusetts Eye & Ear Infirmary Use of azurocidin inhibitors in prevention and treatment of ocular vascular leakage
US8389688B2 (en) 2006-03-06 2013-03-05 Aeres Biomedical, Ltd. Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
CA2647029A1 (en) 2006-04-05 2007-10-18 Abbott Biotechnology Ltd. Antibody purification
WO2007115724A2 (en) * 2006-04-11 2007-10-18 Csl Behring Gmbh Method of increasing the in vivo recovery of therapeutic polypeptides
WO2007124361A2 (en) * 2006-04-20 2007-11-01 Mayo Foundation For Medical Education And Research Soluble b7-h1
AU2007258609B2 (en) * 2006-06-07 2013-01-24 Human Genome Sciences, Inc. Albumin fusion proteins
US7939632B2 (en) 2006-06-14 2011-05-10 Csl Behring Gmbh Proteolytically cleavable fusion proteins with high molar specific activity
PL3896090T3 (en) 2006-06-14 2022-05-02 Csl Behring Gmbh Proteolytically cleavable fusion protein comprising a blood coagulation factor
AU2013202566C1 (en) * 2006-06-14 2018-07-12 Csl Behring Gmbh Proteolytically cleavable fusion protein comprising a blood coagulation factor
EP1867660A1 (en) 2006-06-14 2007-12-19 CSL Behring GmbH Proteolytically cleavable fusion protein comprising a blood coagulation factor
KR101507021B1 (en) * 2006-07-13 2015-03-30 업퍼톤 리미티드 Process for preparing particles of proteinaceous material
EP2046826B1 (en) 2006-07-24 2011-09-14 Biorexis Pharmaceutical Corporation Exendin fusion proteins
JP2009545329A (en) * 2006-08-04 2009-12-24 ファーマシーネ,インコーポレイテッド Long half-life recombinant butyrylcholinesterase
US20110039776A1 (en) * 2006-09-06 2011-02-17 Ashutosh Chilkoti Fusion peptide therapeutic compositions
JP5840345B2 (en) 2006-09-08 2016-01-06 アンブルックス, インコーポレイテッドAmbrx, Inc. Modified human plasma polypeptides or modified human Fc scaffold proteins and uses thereof
US10925977B2 (en) 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
WO2008088422A2 (en) 2006-10-25 2008-07-24 Amgen Inc. Toxin peptide therapeutic agents
CA2666426A1 (en) 2006-10-26 2008-05-02 Novo Nordisk A/S Il-21 variants
CN101636179B (en) 2006-11-07 2012-10-10 默沙东公司 Antagonists of pcsk9
KR20090098880A (en) 2006-12-12 2009-09-17 바이오렉시스 파마슈티칼 코포레이션 Transferrin fusion protein libraries
WO2008074840A2 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the adam family and polypeptides comprising the same for the treatment of adam-related diseases and disorders
WO2008074839A2 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against gpcrs and polypeptides comprising the same for the treatment of gpcr-related diseases and disorders
EP1935430A1 (en) * 2006-12-22 2008-06-25 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
EP3231440A1 (en) 2006-12-22 2017-10-18 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
EP2450371B1 (en) 2007-01-30 2015-04-29 Epivax, Inc. Regulatory t cell epitopes, compositions and uses thereof
EP2109457B1 (en) 2007-02-12 2016-01-06 CSL Behring GmbH Therapeutic application of kazal-type serine protease inhibitors
CA2680832A1 (en) 2007-03-27 2008-10-02 Merck & Co., Inc. Method for detecting autoprocessed, secreted pcsk9
EP2068925A4 (en) 2007-05-07 2011-08-31 Medimmune Llc Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
ES2558689T3 (en) 2007-05-14 2016-02-08 Medimmune, Llc Methods to reduce eosinophil levels
US8420779B2 (en) * 2007-05-22 2013-04-16 Amgen Inc. Compositions and methods for producing bioactive fusion proteins
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
EP2514762B1 (en) 2007-07-13 2015-04-08 The Johns Hopkins University B7-DC variants
AU2008275911A1 (en) * 2007-07-19 2009-01-22 Arizona Board of Regents, a body corporate acting for and on behalf of Arizona State University Self- anchoring MEMS intrafascicular neural electrode
AU2008287340A1 (en) * 2007-08-15 2009-02-19 Amunix, Inc. Compositions and methods for modifying properties of biologically active polypeptides
NZ583605A (en) 2007-08-29 2012-10-26 Sanofi Aventis Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
JP2009055838A (en) * 2007-08-31 2009-03-19 Nipro Corp Fusion protein, gene related to the fusion protein, vector, transformant and anti-inflammatory medicinal composition
CN108129573B (en) * 2007-09-21 2021-10-08 加利福尼亚大学董事会 Targeted interferons exhibit potent apoptotic and antitumor activity
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
CA2701221A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying an fc-containing protein
CA2702448A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying fc-fusion proteins
JP5490714B2 (en) 2007-11-28 2014-05-14 メディミューン,エルエルシー Protein preparation
EP2245064B1 (en) 2007-12-21 2014-07-23 Medimmune Limited BINDING MEMBERS FOR INTERLEUKIN-4 RECEPTOR ALPHA (IL-4Ralpha)
US8092804B2 (en) 2007-12-21 2012-01-10 Medimmune Limited Binding members for interleukin-4 receptor alpha (IL-4Rα)-173
WO2009097318A1 (en) * 2008-01-28 2009-08-06 Bristol-Myers Squibb Company A fluorescence polarization binding assay for characterizing glucokinase ligands
AR070316A1 (en) 2008-02-07 2010-03-31 Merck & Co Inc PCSK9 ANTAGONISTS (SUBTILISINE-KEXINA TYPE 9 PROPROTEIN)
AR070315A1 (en) 2008-02-07 2010-03-31 Merck & Co Inc ANTIBODIES 1B20 ANTAGONISTS OF PCSK9
WO2009111315A2 (en) * 2008-02-29 2009-09-11 Mayo Foundation For Medical Education And Research Methods for reducing granulomatous inflammation
JP5020135B2 (en) * 2008-03-19 2012-09-05 ソニーモバイルコミュニケーションズ, エービー Portable terminal device and computer program
EP2262539B1 (en) 2008-04-01 2015-07-15 Novo Nordisk A/S Insulin albumin conjugates
EP2947097A1 (en) 2008-04-07 2015-11-25 Ablynx N.V. Amino acid sequences directed against the Notch pathways and uses thereof
EP2383292A1 (en) 2008-05-02 2011-11-02 Novartis AG Improved fibronectin-based binding molecules and uses thereof
AU2013202564B2 (en) * 2008-06-24 2015-09-17 Csl Behring Gmbh Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
JP5775452B2 (en) * 2008-07-10 2015-09-09 アンジオン バイオメディカ コーポレイション Compositions and methods of small molecule modulators of hepatocyte growth factor (dispersion factor) activity
CA2731617A1 (en) 2008-07-22 2010-01-28 Maria Joao Saraiva Amino acid sequences directed against multitarget scavenger receptors and polypeptides
DK2328616T3 (en) 2008-08-05 2015-07-20 Novartis Ag Compositions and Methods for Antibodies to Complement Protein C5
PE20110435A1 (en) 2008-08-25 2011-07-20 Amplimmune Inc ANTAGONIST COMPOSITIONS OF PD-1
US20110223188A1 (en) * 2008-08-25 2011-09-15 Solomon Langermann Targeted costimulatory polypeptides and methods of use to treat cancer
US8163497B2 (en) 2008-09-07 2012-04-24 Glyconex Inc. Anti-extended type I glycosphingolipid antibody, derivatives thereof and use
JP5933975B2 (en) 2008-11-12 2016-06-15 メディミューン,エルエルシー Antibody preparation
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
WO2010063011A2 (en) 2008-11-28 2010-06-03 Emory University Methods for the treatment of infections and tumors
BRPI0917592B1 (en) 2008-12-09 2021-08-17 Genentech, Inc ANTI-PD-L1 ANTIBODY, COMPOSITION, MANUFACTURED ARTICLES AND USES OF A COMPOSITION
WO2010068526A1 (en) 2008-12-12 2010-06-17 Merck Sharp & Dohme Corp. Pcsk9 immunoassay
WO2010080833A1 (en) 2009-01-06 2010-07-15 Dyax Corp. Treatment of mucositis with kallikrein inhibitors
WO2010083439A2 (en) 2009-01-16 2010-07-22 Teva Biopharmaceuticals Usa, Inc. Recombinant human albumin-human granulocyte colony stimulating factor for the prevention of neutropenia
WO2010087927A2 (en) 2009-02-02 2010-08-05 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
US8703717B2 (en) * 2009-02-03 2014-04-22 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
PT2393828T (en) 2009-02-03 2017-01-18 Amunix Operating Inc Extended recombinant polypeptides and compositions comprising same
US8680050B2 (en) * 2009-02-03 2014-03-25 Amunix Operating Inc. Growth hormone polypeptides fused to extended recombinant polypeptides and methods of making and using same
SG2014012918A (en) 2009-02-11 2014-04-28 Novozymes Biopharma Dk As Albumin variants and conjugates
US20120121591A1 (en) 2009-03-20 2012-05-17 Amgen Inc. SELECTIVE AND POTENT PEPTIDE INHIBITORS OF Kv1.3
EP2427486B1 (en) 2009-05-07 2015-02-25 Novozymes Biopharma DK A/S Method for purifying albumin
US11512326B2 (en) * 2009-05-26 2022-11-29 University Of Florida Research Foundation, Incorporated Small angiotensin peptide expression system in mammalian cells
EP2258398A1 (en) * 2009-05-26 2010-12-08 Araclón Biotech, S. L. Albumin-amyloid peptide conjugates and uses thereof
US9849188B2 (en) 2009-06-08 2017-12-26 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
AU2010258898B8 (en) 2009-06-08 2015-02-05 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
CN101628346B (en) * 2009-07-25 2012-05-16 大连理工大学 Method for slowing down outlet speed of carbon fiber composite material hand-made hole and buffer device thereof
EP2464661B1 (en) 2009-08-13 2018-01-17 The Johns Hopkins University Methods of modulating immune function with anti-b7-h7cr antibodies
JP6250282B2 (en) 2009-08-20 2017-12-20 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー Albumin fusion clotting factor for non-intravenous administration in the treatment and prophylactic treatment of bleeding disorders
US20110202016A1 (en) * 2009-08-24 2011-08-18 Arsenal Medical, Inc. Systems and methods relating to polymer foams
US10420862B2 (en) 2009-08-24 2019-09-24 Aresenal AAA, LLC. In-situ forming foams for treatment of aneurysms
US9044580B2 (en) 2009-08-24 2015-06-02 Arsenal Medical, Inc. In-situ forming foams with outer layer
JP5813641B2 (en) * 2009-08-24 2015-11-17 アムニクス オペレーティング インコーポレイテッド Coagulation factor IX composition and methods of making and using the same
US9173817B2 (en) 2009-08-24 2015-11-03 Arsenal Medical, Inc. In situ forming hemostatic foam implants
US8451450B2 (en) * 2009-09-14 2013-05-28 Bio-Rad Laboratories, Inc. Near real time optical phase conjugation
UY32917A (en) 2009-10-02 2011-04-29 Boehringer Ingelheim Int DLL-4 BINDING MOLECULES
US20110172398A1 (en) 2009-10-02 2011-07-14 Boehringer Ingelheim International Gmbh Bispecific binding molecules for anti-angiogenesis therapy
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
US20120322075A1 (en) * 2009-10-26 2012-12-20 Externautics S.P.A. Lung Tumor Markers and Methods of Use Thereof
GB2488077A (en) 2009-10-30 2012-08-15 Novozymes Biopharma Dk As Albumin variants
EP2493505A4 (en) 2009-10-30 2013-06-12 Merck Sharp & Dohme Ax1 and ax189 pcsk9 antagonists and variants
CN102639150A (en) 2009-10-30 2012-08-15 默沙东公司 AX213 and AX132 PCSK9 antagonists and variants
WO2011051327A2 (en) 2009-10-30 2011-05-05 Novartis Ag Small antibody-like single chain proteins
WO2011051466A1 (en) 2009-11-02 2011-05-05 Novartis Ag Anti-idiotypic fibronectin-based binding molecules and uses thereof
EP2501714A4 (en) * 2009-11-17 2015-10-07 Univ Montreal Heteropeptides useful for reducing nonspecific adsorption
PL3326643T3 (en) 2009-12-06 2021-10-25 Bioverativ Therapeutics Inc. Factor viii-fc chimeric and hybrid polypeptides, and methods of use thereof
EP2509616A4 (en) 2009-12-08 2013-05-29 Teva Pharma Bche albumin fusions for the treatment of cocaine abuse
US8962807B2 (en) 2009-12-14 2015-02-24 Ablynx N.V. Single variable domain antibodies against OX40L, constructs and therapeutic use
CA2784861A1 (en) 2009-12-21 2011-07-14 Pharmathene, Inc. Recombinant butyrylcholinesterases and truncates thereof
CA3168591A1 (en) 2010-01-06 2011-07-14 Takeda Pharmaceutical Company Limited Plasma kallikrein binding proteins
AR079944A1 (en) 2010-01-20 2012-02-29 Boehringer Ingelheim Int NEUTRALIZING ANTIBODY OF THE ACTIVITY OF AN ANTICOAGULANT
SI2525834T1 (en) 2010-01-22 2019-10-30 Novo Nordisk Healthcare Ag Growth hormones with prolonged in-vivo efficacy
JP5980689B2 (en) 2010-01-22 2016-08-31 ノヴォ・ノルディスク・ヘルス・ケア・アーゲー Stable growth hormone compound
WO2011092233A1 (en) 2010-01-29 2011-08-04 Novartis Ag Yeast mating to produce high-affinity combinations of fibronectin-based binders
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
US8592364B2 (en) * 2010-02-11 2013-11-26 Ecole Polytechnique Federale de Lausanne (“EPFL”) CCR7 ligand delivery and co-delivery in immunotherapy
BR112012022102A2 (en) 2010-03-03 2017-01-10 Boehringer Ingelheim Int a-beta binding polypeptides.
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
KR20130070576A (en) 2010-04-09 2013-06-27 노보자임스 바이오파마 디케이 에이/에스 Albumin derivatives and variants
EP4234698A3 (en) 2010-05-06 2023-11-08 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
KR20130066631A (en) 2010-05-06 2013-06-20 노파르티스 아게 Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
BR112012029588A2 (en) 2010-05-20 2017-07-25 Ablynx Nv her3-related biological materials.
EP3508573A1 (en) 2010-07-09 2019-07-10 Bioverativ Therapeutics Inc. Systems for factor viii processing and methods thereof
NZ605348A (en) 2010-07-09 2015-01-30 Biogen Idec Hemophilia Inc Factor ix polypeptides and methods of use thereof
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9517257B2 (en) 2010-08-10 2016-12-13 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
CA2807942C (en) 2010-08-10 2021-07-27 Ecole Polytechnique Federale De Lausanne Erythrocyte-binding therapeutics
US9850296B2 (en) 2010-08-10 2017-12-26 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
BR112013004012B1 (en) 2010-08-20 2021-03-23 Novartis Ag ISOLATED MONOCLONAL ANTIBODY OR ANTIGEN BINDING FRAGMENT OF THE SAME TO THE HER3 RECEPTOR, ITS USE AND PHARMACEUTICAL COMPOSITION
EP2608807A1 (en) 2010-08-27 2013-07-03 Stem Centrx, Inc. Notum protein modulators and methods of use
EP2611464B1 (en) 2010-09-03 2018-04-25 AbbVie Stemcentrx LLC Novel modulators and methods of use
US20120225081A1 (en) 2010-09-03 2012-09-06 Boehringer Ingelheim International Gmbh Vegf-binding molecules
JP6159660B2 (en) 2010-09-22 2017-07-05 アムジエン・インコーポレーテツド Immunoglobulins as carriers and uses thereof
ES2630031T3 (en) * 2010-09-28 2017-08-17 Aegerion Pharmaceuticals, Inc. A chimeric pinniped-human leptin polypeptide with increased solubility
ES2862955T3 (en) 2010-10-01 2021-10-08 Modernatx Inc Manipulated nucleic acids and methods of using them
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
US8853490B2 (en) * 2010-10-26 2014-10-07 Pioneer Hi Bred International Inc Antifungal proteins and methods of use
US20130225496A1 (en) 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants
DK2635607T3 (en) 2010-11-05 2019-11-18 Zymeworks Inc STABLE HETERODIMED ANTIBODY DESIGN WITH MUTATIONS IN THE FC DOMAIN
KR101650995B1 (en) 2010-11-08 2016-08-25 노파르티스 아게 Cxcr2 binding polypeptides
US9066974B1 (en) 2010-11-13 2015-06-30 Sirbal Ltd. Molecular and herbal combinations for treating psoriasis
US9095606B1 (en) 2010-11-13 2015-08-04 Sirbal Ltd. Molecular and herbal combinations for treating psoriasis
WO2012069466A1 (en) 2010-11-24 2012-05-31 Novartis Ag Multispecific molecules
SG190990A1 (en) 2010-12-08 2013-07-31 Stem Centrx Inc Novel modulators and methods of use
US10393757B2 (en) 2010-12-28 2019-08-27 Dainippon Sumitomo Pharma Co., Ltd. Diagnostic drug and diagnostic method for Alzheimer's disease
US20120171195A1 (en) 2011-01-03 2012-07-05 Ravindranath Mepur H Anti-hla-e antibodies, therapeutic immunomodulatory antibodies to human hla-e heavy chain, useful as ivig mimetics and methods of their use
KR102169651B1 (en) 2011-01-06 2020-10-23 다이액스 코포레이션 Plasma kallikrein binding proteins
SA112330278B1 (en) 2011-02-18 2015-10-09 ستيم سينتركس، انك. Novel modulators and methods of use
WO2012117336A2 (en) 2011-02-28 2012-09-07 Istituto Di Ricovero E Cura A Carattere Scientifico Materno-Infantile Burlo Garofolo - Ospedale Di Alta Specializzazione E Di Rilievo Nazionale Per La Salute Della Donna E Del Bambino Apoptosis-inducing molecules and uses therefor
AU2012222833B2 (en) 2011-03-03 2017-03-16 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
EP2497489A1 (en) 2011-03-09 2012-09-12 CSL Behring GmbH Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
JP6109081B2 (en) 2011-03-09 2017-04-05 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー FXII inhibitors for administration associated with medical procedures involving contact with artificial surfaces
PE20140593A1 (en) 2011-03-16 2014-05-10 Amgen Inc POWERFUL AND SELECTIVE INHIBITORS OF NAV1.3 AND NAV1.7
AR085911A1 (en) 2011-03-16 2013-11-06 Sanofi Sa SAFE THERAPEUTIC DOSE OF A SIMILAR PROTEIN TO AN ANTIBODY WITH VUAL REGION
US9340584B2 (en) 2011-03-29 2016-05-17 The General Hospital Corporation Engineered thioredoxin-like fold proteins
JP2014515740A (en) 2011-03-30 2014-07-03 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Anticoagulant antidote
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US20130078247A1 (en) 2011-04-01 2013-03-28 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to dii4 and ang2
US9527925B2 (en) 2011-04-01 2016-12-27 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to VEGF and ANG2
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
EP2515112B1 (en) * 2011-04-22 2015-08-12 Sysmex Corporation Method for electrochemically detecting analyte
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
GB2491006A (en) * 2011-05-05 2012-11-21 Novozymes Biopharma Uk Ltd Albumin variants
UA117218C2 (en) 2011-05-05 2018-07-10 Мерк Патент Гмбх Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
SI2717898T1 (en) 2011-06-10 2019-07-31 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
US20160145589A1 (en) 2011-06-24 2016-05-26 Green Cross Corporation Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof
RU2727452C1 (en) 2011-06-28 2020-07-21 Инхибркс, Инк. Fused serpin polypeptides and methods for use thereof
AR087020A1 (en) 2011-07-01 2014-02-05 Bayer Ip Gmbh RELAXIN FUSION POLIPEPTIDES AND USES OF THE SAME
CN103649111B (en) 2011-07-05 2018-03-13 阿尔布梅迪克斯医疗公司 Albumin preparation and purposes
PT2729160T (en) 2011-07-08 2019-07-08 Aegerion Pharmaceuticals Inc Engineered polypeptides having enhanced duration of action and reduced immunogenicity
WO2013007563A1 (en) 2011-07-08 2013-01-17 Bayer Intellectual Property Gmbh Fusion proteins releasing relaxin and uses thereof
RU2014103185A (en) 2011-07-18 2015-08-27 Артс Байолоджикс А/С BIOLOGICALLY ACTIVE COMPOUND BASED ON LUTEINIZING HORMONE (LH) WITH PROLONGED ACTION
WO2013014092A1 (en) 2011-07-22 2013-01-31 Csl Behring Gmbh Inhibitory anti -factor xii/xiia monoclonal antibodies and their uses
US20130058947A1 (en) 2011-09-02 2013-03-07 Stem Centrx, Inc Novel Modulators and Methods of Use
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2013052523A1 (en) 2011-10-03 2013-04-11 modeRNA Therapeutics Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US8993831B2 (en) * 2011-11-01 2015-03-31 Arsenal Medical, Inc. Foam and delivery system for treatment of postpartum hemorrhage
EP2773667A1 (en) 2011-11-01 2014-09-10 Bionomics, Inc. Anti-gpr49 antibodies
CA2853951A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Antibodies and methods of treating cancer
US9220774B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of treating cancer by administering anti-GPR49 antibodies
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
WO2013067355A1 (en) 2011-11-04 2013-05-10 Novartis Ag Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
CN109897103A (en) 2011-11-04 2019-06-18 酵活有限公司 There is the antibody design of the stabilization heterodimeric of mutation in Fc structural domain
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
JP2015500829A (en) 2011-12-05 2015-01-08 ノバルティス アーゲー HER3 antibody against domain II of epidermal growth factor receptor 3 (HER3)
CN104114572A (en) 2011-12-16 2014-10-22 现代治疗公司 Modified nucleoside, nucleotide, and nucleic acid compositions
ES2728278T3 (en) 2011-12-21 2019-10-23 Novartis Ag Compositions comprising antibodies directed to factor P and C5
WO2013093027A1 (en) 2011-12-22 2013-06-27 Csl Behring Gmbh Use of c1-inhibitor for the treatment of secondary edema of the central nervous system
US10800847B2 (en) 2012-01-11 2020-10-13 Dr. Mepur Ravindranath Anti-HLA class-IB antibodies mimic immunoreactivity and immunomodulatory functions of intravenous immunoglobulin (IVIG) useful as therapeutic IVIG mimetics and methods of their use
US20130177574A1 (en) 2012-01-11 2013-07-11 Paul I. Terasaki Foundation Laboratory ANTI-HLA CLASS-Ib ANTIBODIES MIMIC IMMUNOREACTIVITY AND IMMUNOMODULATORY FUNCTIONS OF INTRAVENOUS IMMUNOGLOBULIN (IVIg) USEFUL AS THERAPEUTIC IVIg MIMETICS AND METHODS OF THEIR USE
SG11201403764XA (en) 2012-01-12 2014-07-30 Biogen Idec Inc Chimeric factor viii polypeptides and uses thereof
EP2623110A1 (en) 2012-01-31 2013-08-07 CSL Behring GmbH Factor XII inhibitors for the treatment of neurological inflammatory disorders
PL3564260T3 (en) 2012-02-15 2023-03-06 Bioverativ Therapeutics Inc. Factor viii compositions and methods of making and using same
EP2814502B1 (en) * 2012-02-15 2017-09-13 CSL Behring GmbH Von willebrand factor variants having improved factor viii binding affinity
HUE043537T2 (en) 2012-02-15 2019-08-28 Bioverativ Therapeutics Inc Recombinant factor viii proteins
CN110075284A (en) 2012-02-15 2019-08-02 洛桑聚合联合学院 Erythrocyte binding therapeutic agent
ES2812849T3 (en) 2012-02-24 2021-03-18 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and procedures for using them
PE20141859A1 (en) 2012-02-27 2014-12-17 Boehringer Ingelheim Int BINDING POLYEPTIDES TO CX3CR1
US9944691B2 (en) 2012-03-16 2018-04-17 Albumedix A/S Albumin variants
GB201204868D0 (en) * 2012-03-20 2012-05-02 San Raffaele Centro Fond Peptides
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
EP2830646B1 (en) 2012-03-27 2018-03-07 NGM Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
EP2833920A2 (en) 2012-04-02 2015-02-11 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9120853B2 (en) 2012-04-27 2015-09-01 Cytomx Therapeutics, Inc. Activatable antibodies that bind epidermal growth factor receptor and methods of use thereof
WO2013166290A1 (en) 2012-05-04 2013-11-07 Abbvie Biotherapeutics Inc. P21 biomarker assay
US9328174B2 (en) 2012-05-09 2016-05-03 Novartis Ag Chemokine receptor binding polypeptides
WO2013167153A1 (en) 2012-05-09 2013-11-14 Ganymed Pharmaceuticals Ag Antibodies useful in cancer diagnosis
CA2872540A1 (en) 2012-05-10 2013-11-14 Zymeworks Inc. Heteromultimer constructs of immunoglobulin heavy chains with mutations in the fc domain
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
WO2013177386A1 (en) 2012-05-24 2013-11-28 Abbvie Biotherapeutics Inc. Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy
KR102095974B1 (en) 2012-05-30 2020-04-02 바이엘 크롭사이언스 악티엔게젤샤프트 Compositiions comprising a biological control agent and an insecticide
WO2013178656A1 (en) 2012-05-30 2013-12-05 Bayer Cropscience Ag Composition comprising a biological control agent and a fungicide
KR102095979B1 (en) 2012-05-30 2020-04-02 바이엘 크롭사이언스 악티엔게젤샤프트 Compositions comprising a biological control agent and an insecticide
BR112014029122A8 (en) 2012-05-30 2019-09-10 Bayer Cropscience Ag compositions comprising a biological control agent and an insecticide
MX355327B (en) 2012-05-30 2018-04-16 Bayer Cropscience Ag Compositions comprising a biological control agent and a fungicide from the group consisting of inhibitors of the respiratory chain at complex i or ii.
KR102095976B1 (en) 2012-05-30 2020-04-02 바이엘 크롭사이언스 악티엔게젤샤프트 Compositions comprising a biological control agent and an insecticide
CN104507317B (en) 2012-05-30 2019-11-15 拜尔农作物科学股份公司 Composition comprising biocontrol agent and fungicide, and application thereof, kit
PT2854548T (en) 2012-05-30 2018-12-06 Bayer Cropscience Ag Composition comprising a biological control agent and a fungicide selected from metalaxyl and metalaxyl-m
EP3292764A3 (en) 2012-05-30 2018-04-25 Bayer CropScience Aktiengesellschaft Composition comprising a biological control agent and a fungicide selected from inhibitors of the respiratory chain at complex iii
PL2854549T3 (en) 2012-05-30 2019-02-28 Bayer Cropscience Ag Composition comprising a biological control agent and fluopicolide
MX362859B (en) 2012-05-30 2019-02-20 Bayer Cropscience Ag Compositions comprising a biological control agent and an insecticide.
EP2854546B1 (en) 2012-05-30 2018-07-04 Bayer Cropscience AG Composition comprising a biological control agent and a fungicide selected from inhibitors of the ergosterol biosynthesis
EP3693000B1 (en) 2012-06-08 2022-03-02 Bioverativ Therapeutics Inc. Procoagulant compounds
CA2875247A1 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
KR101380740B1 (en) 2012-06-29 2014-04-11 쉐어 휴먼 제네텍 세러피스, 인코포레이티드 Purification of iduronate-2-sulfatase
US9150841B2 (en) 2012-06-29 2015-10-06 Shire Human Genetic Therapies, Inc. Cells for producing recombinant iduronate-2-sulfatase
US10656156B2 (en) 2012-07-05 2020-05-19 Mepur Ravindranath Diagnostic and therapeutic potential of HLA-E monospecific monoclonal IgG antibodies directed against tumor cell surface and soluble HLA-E
NZ703366A (en) 2012-07-11 2018-03-23 Amunix Operating Inc Factor viii complex with xten and von willebrand factor protein, and uses thereof
WO2014012082A2 (en) 2012-07-13 2014-01-16 Zymeworks Inc. Multivalent heteromultimer scaffold design an constructs
US20150202287A1 (en) 2012-08-30 2015-07-23 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-erbb3 agents
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
SG11201504249XA (en) 2012-09-02 2015-07-30 Abbvie Inc Methods to control protein heterogeneity
WO2014063108A1 (en) 2012-10-18 2014-04-24 Biogen Idec Ma Inc. Methods of using a fixed dose of a clotting factor
BR112015010318A2 (en) 2012-11-08 2017-08-22 Albumedix As ALBUMIN VARIANTS
KR20240033145A (en) 2012-11-13 2024-03-12 비온테크 에스이 Agents for treatment of claudin expressing cancer diseases
PL2922554T3 (en) 2012-11-26 2022-06-20 Modernatx, Inc. Terminally modified rna
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
EP3851454A1 (en) 2012-12-05 2021-07-21 Novartis AG Compositions and methods for antibodies targeting epo
EP2935311B1 (en) 2012-12-20 2021-03-31 Amgen Inc. Apj receptor agonists and uses thereof
CA2896793A1 (en) 2013-01-15 2014-07-24 Teva Pharmaceutical Industries Ltd. Formulations of albu-bche, preparation and uses thereof
KR101503907B1 (en) * 2013-01-17 2015-03-20 서울대학교산학협력단 Recombinant protein vaccine for preventing Actinobacillus pleuropneumoniae and Mycoplasma hyopneumoniae infection diseases
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
US9161966B2 (en) 2013-01-30 2015-10-20 Ngm Biopharmaceuticals, Inc. GDF15 mutein polypeptides
EP2953469A1 (en) 2013-02-11 2015-12-16 Bayer Cropscience LP Compositions comprising a streptomyces-based biological control agent and another biological control agent
PL2956477T3 (en) 2013-02-15 2021-06-14 Bioverativ Therapeutics Inc. Optimized factor viii gene
EP2956002B1 (en) 2013-02-16 2017-09-06 Albumedix A/S Pharmacokinetic animal model
WO2014131865A1 (en) 2013-02-28 2014-09-04 Csl Behring Gmbh Therapeutic agent for amniotic fluid embolism
US9255262B2 (en) * 2013-03-06 2016-02-09 Vision Global Holdings Ltd. Albumin-binding arginine deminase and the use thereof
USRE48805E1 (en) 2013-03-06 2021-11-02 Vision Global Holdings Ltd. Method for cancer targeting treatment and detection of arginine using albumin-binding arginine deiminase fusion protein
AU2014224599B2 (en) 2013-03-08 2018-11-08 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
CA2940513C (en) 2013-03-11 2023-08-15 University Of Florida Research Foundation, Inc. Delivery of card protein as therapy for ocular inflammation
EP2830651A4 (en) 2013-03-12 2015-09-02 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
UY35397A (en) 2013-03-12 2014-10-31 Amgen Inc POWERFUL AND SELECTIVE INHIBITORS OF NaV1.7
US20160024181A1 (en) * 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US20160152686A1 (en) 2013-03-13 2016-06-02 Bristol-Myers Squibb Company Fibronectin based scaffold domains linked to serum albumin or moiety binding thereto
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
EP3611189A1 (en) 2013-03-14 2020-02-19 Novartis AG Antibodies against notch 3
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
SG10201913874TA (en) 2013-03-15 2020-03-30 Biogen Ma Inc Factor ix polypeptide formulations
EP3473272A1 (en) 2013-03-29 2019-04-24 Alexion Pharmaceuticals, Inc. Compositions and methods for increasing the serum half-life of a therapeutic agent targeting complement c5
WO2014166836A1 (en) * 2013-04-05 2014-10-16 Novo Nordisk A/S Growth hormone compound formulation
ES2657291T3 (en) 2013-04-22 2018-03-02 Csl Ltd. A covalent complex of von Willebrand factor and factor VIII associated by a disulfide bridge
AR096601A1 (en) 2013-06-21 2016-01-20 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
UY35620A (en) 2013-06-21 2015-01-30 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
PL3013366T3 (en) 2013-06-28 2022-01-17 Csl Behring Gmbh Combination therapy using a factor xii inhibitor and a c1-inhibitor
CN104342420B (en) * 2013-07-30 2017-09-15 惠觅宙 A kind of recombinant long-acting people hyaluronidase, its encoding gene, production method and application
TW202003554A (en) 2013-08-14 2020-01-16 美商百歐維拉提夫治療公司 Factor VIII-XTEN fusions and uses thereof
EP3033098B1 (en) 2013-08-14 2022-06-22 Bioverativ Therapeutics Inc. Recombinant factor viii proteins
US9387151B2 (en) 2013-08-20 2016-07-12 Anutra Medical, Inc. Syringe fill system and method
CN105792836A (en) 2013-08-28 2016-07-20 施特姆森特克斯股份有限公司 Novel SEZ6 modulators and methods of use
AU2014312215B2 (en) 2013-08-28 2020-02-27 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
ES2900425T3 (en) 2013-09-25 2022-03-16 Bioverativ Therapeutics Inc Column viral inactivation methods
CN103468662A (en) * 2013-09-29 2013-12-25 惠觅宙 Recombined human hyaluronidase, production and purification method and preparations thereof, use method and application
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015051293A2 (en) 2013-10-04 2015-04-09 Abbvie, Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
WO2015057403A2 (en) * 2013-10-17 2015-04-23 Tarix Pharmaceuticals Ltd. Compositions and methods for treatment of inflammatory bowel disease
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9540440B2 (en) 2013-10-30 2017-01-10 Cytomx Therapeutics, Inc. Activatable antibodies that bind epidermal growth factor receptor and methods of use thereof
WO2015066550A1 (en) 2013-10-31 2015-05-07 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
JP6306700B2 (en) * 2013-11-01 2018-04-04 ユニバーシティ オブ オスロUniversity of Oslo Modified albumin and use thereof
EP3065769A4 (en) 2013-11-08 2017-05-31 Biogen MA Inc. Procoagulant fusion compound
US20150139988A1 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
WO2015089283A1 (en) 2013-12-11 2015-06-18 Cytomx Therapeutics, Inc. Antibodies that bind activatable antibodies and methods of use thereof
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
CA2935903A1 (en) 2014-01-09 2015-07-16 Hadasit Medical Research Services And Development Ltd. Improved cell compositions and methods for cancer therapy
CN117106095A (en) 2014-01-10 2023-11-24 比奥贝拉蒂治疗公司 Factor VIII chimeric proteins and uses thereof
US10046056B2 (en) 2014-02-21 2018-08-14 École Polytechnique Fédérale De Lausanne (Epfl) Glycotargeting therapeutics
US10946079B2 (en) 2014-02-21 2021-03-16 Ecole Polytechnique Federale De Lausanne Glycotargeting therapeutics
SG10202010936RA (en) 2014-02-21 2020-12-30 Ecole Polytecnique Fed De Lausanne Epfl Epfl Tto Glycotargeting therapeutics
US10953101B2 (en) 2014-02-21 2021-03-23 École Polytechnique Fédérale De Lausanne (Epfl) Glycotargeting therapeutics
ES2911714T3 (en) 2014-03-11 2022-05-20 Univ Florida AAV-expressed protein M013 as an anti-inflammatory therapeutic for use in a method of treating inflammatory eye disease
US20170107294A1 (en) * 2014-03-21 2017-04-20 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
WO2015160618A1 (en) 2014-04-16 2015-10-22 Bayer Cropscience Lp Compositions comprising ningnanmycin and a biological control agent
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
MX2016014306A (en) 2014-05-02 2017-06-12 Cerenis Therapeutics Holding Sa Hdl therapy markers.
WO2015179654A1 (en) 2014-05-22 2015-11-26 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti b7-h1 antibodies
CN105198999A (en) * 2014-05-27 2015-12-30 上海生物制品研究所有限责任公司 Fusion protein and its preparation method and use
USD750768S1 (en) 2014-06-06 2016-03-01 Anutra Medical, Inc. Fluid administration syringe
USD774182S1 (en) 2014-06-06 2016-12-13 Anutra Medical, Inc. Anesthetic delivery device
USD763433S1 (en) 2014-06-06 2016-08-09 Anutra Medical, Inc. Delivery system cassette
CA2951391C (en) 2014-06-10 2021-11-02 Amgen Inc. Apelin polypeptides
CA2950988C (en) 2014-06-18 2024-02-06 Csl Behring Gmbh Therapy using a factor xii inhibitor in a neurotraumatic disorder
US20170204149A1 (en) 2014-06-23 2017-07-20 Novartis Ag Hsa-gdf-15 fusion polypeptide and use thereof
TW201613958A (en) 2014-06-24 2016-04-16 Novo Nordisk As MIC-1 fusion proteins and uses thereof
EP3160478A4 (en) 2014-06-30 2018-05-16 Bioverativ Therapeutics Inc. Optimized factor ix gene
WO2016014148A1 (en) 2014-07-23 2016-01-28 Mayo Foundation For Medical Education And Research Targeting dna-pkcs and b7-h1 to treat cancer
PE20170771A1 (en) 2014-07-30 2017-07-04 Ngm Biopharmaceuticals Inc COMPOSITIONS AND METHODS OF USE TO TREAT METABOLIC DISORDERS
DK3177642T3 (en) 2014-08-07 2022-02-21 Novartis Ag ANGIOPOIETIN-LIKE 4 ANTIBODIES AND METHODS OF USING IT
EP3194437B1 (en) 2014-08-07 2021-01-20 Novartis AG Angiopoietin-like 4 (angptl4) antibodies and methods of use
MX2017002323A (en) 2014-08-22 2017-08-02 Univ Nat Cheng Kung Disintegrin variants and pharmaceutical uses thereof.
AR101956A1 (en) 2014-09-17 2017-01-25 Bayer Cropscience Lp COMPOSITIONS THAT INCLUDE BACILLUS RECOMBINATING CELLS AND ANOTHER BIOLOGICAL CONTROL AGENT
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
PE20170702A1 (en) 2014-09-26 2017-06-24 Bayer Pharma AG STABILIZED DERIVATIVES OF ADRENOMEDULIN AND THE USE OF THEM
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
BR112017007765B1 (en) 2014-10-14 2023-10-03 Halozyme, Inc COMPOSITIONS OF ADENOSINE DEAMINASE-2 (ADA2), VARIANTS THEREOF AND METHODS OF USING THE SAME
MA40835A (en) 2014-10-23 2017-08-29 Biogen Ma Inc ANTI-GPIIB / IIIA ANTIBODIES AND THEIR USES
EP3701969A1 (en) 2014-10-31 2020-09-02 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
US20160130324A1 (en) * 2014-10-31 2016-05-12 Shire Human Genetic Therapies, Inc. C1 Inhibitor Fusion Proteins and Uses Thereof
GB201420139D0 (en) 2014-11-12 2014-12-24 Ucl Business Plc Factor IX gene therapy
UY36449A (en) 2014-12-19 2016-07-29 Novartis Ag COMPOSITIONS AND METHODS FOR ANTIBODIES DIRECTED TO BMP6
TWI669129B (en) * 2014-12-23 2019-08-21 德商梅茲製藥有限兩合公司 Refilled glass container, a kit comprising the same and, a use thereof
WO2016161010A2 (en) * 2015-03-30 2016-10-06 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to fc gamma receptors
JP2016204292A (en) * 2015-04-21 2016-12-08 国立大学法人 熊本大学 Bmp7 mutant-albumin cointegrate, and renal disease therapeutic agent containing the same
KR20170138569A (en) * 2015-04-29 2017-12-15 메디오라늄 파마세우티시 에스.피.에이. Soluble chimeric interleukin-10 receptor and its therapeutic use
WO2016195723A1 (en) * 2015-06-03 2016-12-08 Beller Pharmaceuticals LLC Methods of treatment for conditions mediated by substance p
PE20180041A1 (en) 2015-06-05 2018-01-09 Novartis Ag ANTIBODIES TARGETING BONE MORPHOGENETIC PROTEIN (BMP9) AND METHODS FROM THESE
CA2989110A1 (en) * 2015-06-11 2016-12-15 Attwill Medical Solutions Inc. Medical devices, systems, and methods utilizing antithrombin-heparin compositions
JOP20200312A1 (en) 2015-06-26 2017-06-16 Novartis Ag Factor xi antibodies and methods of use
ES2852002T3 (en) * 2015-07-30 2021-09-10 Endor Tech S L Colony stimulating factor for use in the treatment of colon or pancreatic cancer
EP3331914A1 (en) 2015-08-03 2018-06-13 Novartis AG Methods of treating fgf21-associated disorders
BR112018002150A2 (en) 2015-08-03 2018-09-18 Bioverativ Therapeutics Inc factor ix fusion proteins and methods of manufacturing and using them
AU2016302335B2 (en) * 2015-08-06 2022-08-04 The Trustees Of The University Of Pennsylvania GLP-1 and use thereof in compositions for treating metabolic diseases
JP7007261B2 (en) 2015-08-20 2022-01-24 アルブミディクス リミティド Albumin variants and conjugates
JP6886920B2 (en) 2015-09-08 2021-06-16 Jcrファーマ株式会社 New human serum albumin mutant
TN2018000076A1 (en) 2015-09-09 2019-07-08 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding molecules and methods of using the molecules
DK3347377T3 (en) 2015-09-09 2021-05-10 Novartis Ag Thymic stromal lymphopoietin (TSLP) -binding antibodies and methods of using the antibodies
CN108430518A (en) 2015-09-11 2018-08-21 小利兰·斯坦福大学托管委员会 Biological dependent quadrature cell factor/receptor pair
WO2017075045A2 (en) 2015-10-30 2017-05-04 Mayo Foundation For Medical Education And Research Antibodies to b7-h1
KR20180081825A (en) 2015-12-04 2018-07-17 베링거 인겔하임 인터내셔날 게엠베하 Biparotopic polypeptides that antagonize WNT signaling in tumor cells
WO2017100679A1 (en) 2015-12-11 2017-06-15 Dyax Corp. Plasma kallikrein inhibitors and uses thereof for treating hereditary angioedema attack
UY37030A (en) 2015-12-18 2017-07-31 Novartis Ag ANTIBODIES DIRECTED TO CD32B AND METHODS OF USE OF THE SAME
CN108367053A (en) 2015-12-22 2018-08-03 诺华股份有限公司 The method that metabolic disease is treated or improved using growth and differentiation factor 15 (GDF-15)
CN116003593A (en) 2016-01-11 2023-04-25 苏黎世大学 Immunostimulatory humanized monoclonal antibodies directed against human interleukin-2 and fusion proteins thereof
ES2847155T3 (en) 2016-01-21 2021-08-02 Novartis Ag Multispecific molecules targeting CLL-1
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
JP2017165713A (en) 2016-03-14 2017-09-21 Jcrファーマ株式会社 Serum albumin-20K growth hormone fusion protein
CA3018081A1 (en) 2016-03-22 2017-09-28 Bionomics Limited Administration of an anti-lgr5 monoclonal antibody
KR102370762B1 (en) 2016-03-31 2022-03-04 엔지엠 바이오파마슈티컬스, 아이엔씨. Binding proteins and methods of use thereof
EP3440107A4 (en) 2016-04-06 2020-02-19 CSL Limited Method of treating atherosclerosis
JP7138567B2 (en) 2016-04-27 2022-09-16 ノバルティス アーゲー Antibodies against growth differentiation factor 15 and their uses
EP3848393A1 (en) 2016-05-18 2021-07-14 Boehringer Ingelheim International GmbH Antibody molecules for cancer treatment
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
WO2018007601A1 (en) * 2016-07-08 2018-01-11 Csl Behring Recombinant Facility Ag Subcutaneous administration of long-acting factor ix in humans
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
AU2017305856A1 (en) 2016-08-05 2019-03-21 Csl Behring Gmbh Pharmaceutical formulations of C1 esterase inhibitor
EP3504648A1 (en) 2016-08-23 2019-07-03 CSL Behring GmbH Method of preventing acute attacks of hereditary angioedema associated with c1 esterase inhibitor deficiency
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
JP2020500306A (en) 2016-11-04 2020-01-09 オーフス ウニベルシテット Identification and treatment of tumors characterized by neonatal Fc receptor overexpression
US11324804B2 (en) * 2016-11-18 2022-05-10 Sepsia Therapeutics, S.L. Combined CD6 and imipenem therapy for treatment of infectious diseases and related inflammatory processes
WO2018096396A1 (en) 2016-11-22 2018-05-31 University Of Oslo Albumin variants and uses thereof
CN110520150A (en) 2016-12-02 2019-11-29 比奥维拉迪维治疗股份有限公司 Use the method for chimeric coagulation factor therapies hemophilic arthosis
AU2017368328A1 (en) 2016-12-02 2019-07-18 Bioverativ Therapeutics Inc. Methods of inducing immune tolerance to clotting factors
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
MA47163A (en) 2016-12-16 2019-11-06 Biogen Ma Inc PROTEOLYTICALLY STABILIZED ACTIVATED GROWTH DIFFERENTIATION FACTOR 11
EP3360898A1 (en) 2017-02-14 2018-08-15 Boehringer Ingelheim International GmbH Bispecific anti-tnf-related apoptosis-inducing ligand receptor 2 and anti-cadherin 17 binding molecules for the treatment of cancer
EP3558370A2 (en) 2016-12-23 2019-10-30 Novartis AG Methods of treatment with anti-factor xi/xia antibodies
IL267538B1 (en) 2016-12-23 2024-01-01 Novartis Ag Anti-factor xi/xia antibodies for use in preventing, treating, managing or reducing the risk of a thromboembolic disorder or stroke in a subject
GB2605925B (en) 2016-12-23 2023-02-22 Harvard College Gene editing of PCSK9
US20190382462A1 (en) 2017-01-13 2019-12-19 Pietro P. Sanna Methods and compositions for treating hpa hyperactivity
EA201991768A1 (en) 2017-01-31 2020-01-22 Байоверетив Терапьютикс Инк. FUSIONED PROTEINS BASED ON FACTOR IX AND METHODS OF PRODUCING THEM AND WAYS OF APPLICATION
MX2019009498A (en) 2017-02-08 2019-10-02 Novartis Ag Fgf21 mimetic antibodies and uses thereof.
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
US20200131225A1 (en) 2017-04-20 2020-04-30 Novo Nordisk A/S Methods of purification of albumin fusion proteins
TWI710377B (en) 2017-05-23 2020-11-21 丹麥商諾佛 儂迪克股份有限公司 Mic-1 compounds and uses thereof
KR20200011933A (en) 2017-05-31 2020-02-04 베링거 인겔하임 인터내셔날 게엠베하 Polypeptides that antagonize WNT signaling in tumor cells
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2018232176A1 (en) 2017-06-16 2018-12-20 The University Of Chicago Compositions and methods for inducing immune tolerance
KR102597725B1 (en) 2017-06-29 2023-11-06 체에스엘 베링 렝나우 아게 21-day dosing regimen and method for fusion protein comprising factor IX and human albumin for prophylactic treatment of hemophilia
AU2018309090B2 (en) 2017-08-04 2023-03-30 Amgen Inc. Method of conjugation of cys-mAbs
CA3072334A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
EP3684811A2 (en) 2017-08-17 2020-07-29 Massachusetts Institute of Technology Multiple specificity binders of cxc chemokines and uses thereof
EP3681517A4 (en) 2017-09-15 2021-05-19 Cedars-Sinai Medical Center Methods for improving organ function in organ transplant patients
CN111247165B (en) 2017-10-18 2023-11-10 Csl有限公司 Human serum albumin variants and uses thereof
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
CA3085478A1 (en) 2017-12-15 2019-06-20 Csl Limited Use of a fxiia-inhibitor in the treatment of renal fibrosis and/or chronic kidney disease
WO2019129054A1 (en) 2017-12-27 2019-07-04 信达生物制药(苏州)有限公司 Triabody, preparation method and use thereof
EP3732202A4 (en) 2017-12-28 2022-06-15 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against tigit
US11246908B2 (en) * 2018-01-10 2022-02-15 The Johns Hopkins University Compositions comprising albumin-FMS-like tyrosine kinase 3 ligand fusion proteins and uses thereof
WO2019137541A1 (en) 2018-01-15 2019-07-18 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against pd-1
JP2021512126A (en) 2018-02-01 2021-05-13 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 Use of a lentiviral vector expressing factor VIII
JP2021518426A (en) * 2018-03-13 2021-08-02 ホスピタル クリニック デ バルセロナ Bacterial binding peptides for the treatment of infectious diseases and related inflammatory processes
JP2021515793A (en) 2018-03-13 2021-06-24 ザイムワークス,インコーポレイテッド Anti-HER2 Biparatopic Antibody-Drug Conjugate and Usage
CN112154153A (en) 2018-03-28 2020-12-29 百时美施贵宝公司 Interleukin-2/interleukin-2 receptor alpha fusion proteins and methods of use
CN111886254B (en) 2018-03-30 2023-12-08 南京传奇生物科技有限公司 Single domain antibodies against LAG-3 and uses thereof
KR102119197B1 (en) * 2018-04-23 2020-06-05 주식회사 엘베이스 Composition for autophagy inhibiting in cell, And pharmaceutical composition for preventing or treating tumor disease, or inhibiting anti-cancer agents resistance containing the same
KR20210021467A (en) 2018-05-14 2021-02-26 웨어울프 세라퓨틱스, 인크. Activatable interleukin-2 polypeptide and method of use thereof
BR112020023118A2 (en) 2018-05-14 2021-04-13 Werewolf Therapeutics, Inc. ACTIVE ACTIVABLE INTERLEUKIN POLYPEPTIDS 12 AND METHODS OF USE OF THESE
US20190375822A1 (en) 2018-05-18 2019-12-12 Bioverativ Therapeutics Inc. Methods of treating hemophilia a
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
TW202015726A (en) 2018-05-30 2020-05-01 瑞士商諾華公司 Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
EP3802611A2 (en) 2018-06-01 2021-04-14 Novartis AG Binding molecules against bcma and uses thereof
EP3817720A2 (en) 2018-07-03 2021-05-12 Bristol-Myers Squibb Company Fgf21 formulations
CN113227385A (en) 2018-08-09 2021-08-06 比奥维拉迪维治疗股份有限公司 Nucleic acid molecules and their use for non-viral gene therapy
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
JP2021535142A (en) 2018-08-31 2021-12-16 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Dosing strategies to reduce cytokine release syndrome of CD3 / C20 bispecific antibodies
EP3852874A1 (en) * 2018-09-17 2021-07-28 Kyoto University Administration of an anti-c5 agent for treatment of hepatic injury or failure
AU2019347751A1 (en) 2018-09-27 2021-04-29 Xilio Development, Inc. Masked cytokine polypeptides
UY38407A (en) 2018-10-15 2020-05-29 Novartis Ag TREM2 STABILIZING ANTIBODIES
US20210388059A1 (en) * 2018-10-29 2021-12-16 Spin Therapeutics, Llc Compositions and methods for alpha-1-antitrypsin disorders
GB201818477D0 (en) 2018-11-13 2018-12-26 Emstopa Ltd Tissue plasminogen activator antibodies and method of use thereof
WO2020109978A1 (en) 2018-11-26 2020-06-04 Novartis Ag Lpl-gpihbp1 fusion polypeptides
CA3121786A1 (en) 2018-12-06 2020-06-11 Bioverativ Therapeutics Inc. Use of lentiviral vectors expressing factor ix
EP3941580A1 (en) 2019-03-22 2022-01-26 Reflexion Pharmaceuticals, Inc. D-peptidic compounds for vegf
US20230051872A1 (en) 2019-03-22 2023-02-16 Reflexion Pharmaceuticals, Inc. Multivalent D-Peptidic Compounds for Target Proteins
CN113874392A (en) 2019-03-28 2021-12-31 丹尼斯科美国公司 Engineered antibodies
MX2021012250A (en) 2019-04-11 2022-01-18 Angion Biomedica Corp Solid forms of (e)-3-[2-(2-thienyl)vinyl]-1h-pyrazole.
CA3137512A1 (en) 2019-05-14 2020-11-19 Werewolf Therapeutics, Inc. Separation moieties and methods and use thereof
EP3972993A1 (en) 2019-05-21 2022-03-30 Novartis AG Variant cd58 domains and uses thereof
EP3972998A1 (en) 2019-05-21 2022-03-30 Novartis AG Cd19 binding molecules and uses thereof
JP2022532928A (en) 2019-05-24 2022-07-20 サノフイ Methods for treating systemic scleroderma
CA3143584A1 (en) 2019-06-18 2020-12-24 Bayer Aktiengesellschaft Adrenomedullin-analogues for long-term stabilization and their use
US20230308835A1 (en) 2019-09-06 2023-09-28 Novartis Ag Therapeutic fusion proteins
US20220348651A1 (en) 2019-09-18 2022-11-03 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
US20210113634A1 (en) 2019-09-30 2021-04-22 Bioverativ Therapeutics Inc. Lentiviral vector formulations
EP4047006A4 (en) 2019-10-17 2023-11-01 JCR Pharmaceuticals Co., Ltd. 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
EP4069200A1 (en) 2019-12-04 2022-10-12 Albumedix Ltd Methods and compositions produced thereby
AU2020401371A1 (en) 2019-12-13 2022-07-21 Synthekine, Inc. IL-2 orthologs and methods of use
US20210181200A1 (en) * 2019-12-17 2021-06-17 Women's College Hospital Ovarian cancer biomarker and methods of using same
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
WO2021236658A1 (en) 2020-05-19 2021-11-25 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
EP4232474A1 (en) 2020-10-21 2023-08-30 Boehringer Ingelheim International GmbH Bispecific anti-vegf and anti-trkb binding molecules for the treatment of eye diseases
EP4240491A1 (en) 2020-11-06 2023-09-13 Novartis AG Cd19 binding molecules and uses thereof
WO2022166720A1 (en) * 2021-02-05 2022-08-11 华南理工大学 Serum albumin-based fusion protein, and nano-assembly, preparation method therefor and application thereof
EP4294834A2 (en) 2021-02-19 2023-12-27 The United States of America, as represented by The Secretary, Department of Health and Human Services Single domain antibodies that neutralize sars-cov-2
US20220411533A1 (en) 2021-06-17 2022-12-29 Boehringer Ingelheim International Gmbh Novel Tri-specific Binding Molecules
CN114133458B (en) * 2021-12-08 2023-11-14 福州大学 Method for fusing polypeptide in human serum albumin
TW202400658A (en) 2022-04-26 2024-01-01 瑞士商諾華公司 Multispecific antibodies targeting il-13 and il-18
WO2024013315A1 (en) 2022-07-15 2024-01-18 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies

Family Cites Families (517)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
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
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
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
US4447538A (en) * 1978-04-19 1984-05-08 Regents Of The University Of California Microorganism containing gene for human chorionic somatomammotropin
US5326859A (en) 1979-10-30 1994-07-05 Juridical Foundation, Japanese Foundation For Cancer Research DNA and recombinant plasmid
US5514567A (en) * 1979-01-30 1996-05-07 Juridical Foundation, Japanese Foundation For Cancer Research DNA and recombinant plasmid
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
US4530901A (en) 1980-01-08 1985-07-23 Biogen N.V. Recombinant DNA molecules and their use in producing human interferon-like polypeptides
DK170236B1 (en) 1980-01-08 1995-07-10 Biogen Inc Recombinant DNA molecules, method for preparing an IFN-alpha type polypeptide and method for selecting DNA sequences encoding IFN-alpha type polypeptides
ES8302096A1 (en) 1980-04-03 1982-12-16 Biogen Nv DNA sequences, recombinant DNA molecules and processes for producing human fibroblast interferon.
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
US4456748A (en) 1981-02-23 1984-06-26 Genentech, Inc. Hybrid human leukocyte interferons
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
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4792602A (en) 1981-06-19 1988-12-20 Cornell Research Foundation, Inc. Adaptors, and synthesis and cloning of proinsulin genes
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
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
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
WO1983002461A1 (en) 1982-01-19 1983-07-21 Cetus Corp Multiclass hybrid interferons
EP0088046B1 (en) 1982-02-17 1987-12-09 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
US4670393A (en) * 1982-03-22 1987-06-02 Genentech, Inc. DNA vectors encoding a novel human growth hormone-variant protein
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
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
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
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
FI82266C (en) 1982-10-19 1991-02-11 Cetus Corp Process for Preparation of IL-2 Mutein
US4966843A (en) * 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
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
NZ207394A (en) 1983-03-08 1987-03-06 Commw Serum Lab Commission Detecting or determining sequence of amino acids
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
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4518584A (en) 1983-04-15 1985-05-21 Cetus Corporation Human recombinant interleukin-2 muteins
JPS60501140A (en) 1983-04-22 1985-07-25 アムジエン Secretion of exogenous polypeptides by 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
US4576813A (en) 1983-07-05 1986-03-18 Monsanto Company Heat recovery from concentrated sulfuric acid
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
US4518564A (en) * 1983-10-03 1985-05-21 Jeneric Industries, Inc. Gallium and silver free, palladium based dental alloys for porcelain-fused-to-metal restorations
EP0143949B1 (en) 1983-11-01 1988-10-12 TERUMO KABUSHIKI KAISHA trading as TERUMO CORPORATION Pharmaceutical composition containing urokinase
GB8334102D0 (en) 1983-12-21 1984-02-01 Searle & Co Interferons with cysteine pattern
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
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
JPS60136596A (en) 1983-12-26 1985-07-20 Suntory Ltd Peptide and diuretic comprising it as active ingredient
US4908434A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Process for preparing purified interleukin-2
US4908433A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Uses of 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
FR2564106B1 (en) * 1984-05-09 1988-04-22 Transgene Sa FACTOR IX EXPRESSION VECTORS, CELLS TRANSFORMED BY THESE VECTORS, AND PROCESS FOR THE PREPARATION OF FACTOR IX.
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
US5908763A (en) 1984-07-06 1999-06-01 Novartis Corporation DNA encoding GM-CSF and a method of producing GM-CSF protein
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
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
US4734491A (en) 1984-08-31 1988-03-29 University Patents, Inc. DNA sequences encoding hybrid lymphoblastoid-leukocyte human interferons
US4716217A (en) 1984-08-31 1987-12-29 University Patents, Inc. Hybrid lymphoblastoid-leukocyte human interferons
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
US4959314A (en) * 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
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
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
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.
JP2532858B2 (en) 1985-04-01 1996-09-11 セルテツク リミテツド Transformed myeloma cell line
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
GB8510219D0 (en) 1985-04-22 1985-05-30 Bass Plc Isolation of fermentation products
AU5890286A (en) 1985-06-17 1986-12-24 Genex Corp. Cloned human serum albumin gene
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
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
DE3689123T2 (en) 1985-11-01 1994-03-03 Xoma Corp MODULAR UNIT OF ANTIBODY GENES, ANTIBODIES MADE THEREOF AND USE.
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
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
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
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
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
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
IT1204400B (en) * 1986-06-20 1989-03-01 Sclavo Spa PHARMACEUTICAL COMPOSITIONS CONTAINING A CALCITONIN
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
JPH01502669A (en) 1987-03-13 1989-09-14 アムジエン・インコーポレーテツド Purified platelet-derived growth factor and its purification method
US4835260A (en) * 1987-03-20 1989-05-30 Genetics Institute, Inc. Erythropoietin composition
EP0286424B1 (en) * 1987-04-09 1994-03-16 Delta Biotechnology Limited Yeast vector
JPH0198489A (en) 1987-04-22 1989-04-17 Chiron Corp Recombinant production of platelet-derived growth factor a chain polypeptide
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US4994560A (en) 1987-06-24 1991-02-19 The Dow Chemical Company Functionalized polyamine chelants and radioactive rhodium complexes thereof for conjugation to antibodies
US5489425A (en) 1987-06-24 1996-02-06 The Dow Chemical Company Functionalized polyamine chelants
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
FI107939B (en) 1987-07-28 2001-10-31 Dsm Nv Kluyveromyces as a host strain
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
NZ226414A (en) 1987-10-02 1992-07-28 Genentech Inc Cd4 peptide adhesion variants and their preparation and use
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
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
ZA89430B (en) 1988-01-22 1989-10-25 Gen Hospital Corp Cloned genes encoding ig-cd4 fusion proteins and the use thereof
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
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
US5763394A (en) * 1988-04-15 1998-06-09 Genentech, Inc. Human growth hormone aqueous 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
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
US5096885A (en) * 1988-04-15 1992-03-17 Genentech, Inc. Human growth hormone formulation
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
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
US5756065A (en) 1988-06-24 1998-05-26 The Dow Chemical Company Macrocyclic tetraazacyclododecane conjugates and their use as diagnostic and therapeutic agents
ZA894792B (en) 1988-06-24 1991-04-24 Dow Chemical Co Macrocyclic bifunctional chelants,complexes thereof and their antibody conjugates
HU219485B (en) 1988-06-24 2001-04-28 Dow Chemical Co. Process for producing 1,4,7,10-tetraazacyclododecane derivatives, complexes thereof, their antibody conjugates and pharmaceutical compositions containing them and diagnostic compositions containing the complexes and the conjugates
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
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
FR2635115B1 (en) 1988-08-05 1992-04-03 Rhone Poulenc Sante PROCESS FOR THE PREPARATION OF HUMAN ALBUMIN SERUM FROM YEAST
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
DE68927933T2 (en) 1988-09-02 1997-08-14 Dyax Corp PRODUCTION AND SELECTION OF RECOMBINANT PROTEINS WITH DIFFERENT BINDING POINTS
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
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
US5298243A (en) 1988-10-20 1994-03-29 Denki Kagaku Kogyo Kabushiki Kaisha Colony stimulating factor-gelatin conjugate
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
US5696239A (en) 1988-10-31 1997-12-09 The Dow Chemical Company Conjugates possessing ortho ligating functionality and complexes thereof
US5342604A (en) 1988-10-31 1994-08-30 The Dow Chemical Company Complexes possessing ortho ligating functionality
US5256410A (en) 1988-12-01 1993-10-26 Schering Corporation Treatment of squamous cell carcinoma intralesionally with recombinant human alpha interferon
US4975271A (en) 1988-12-19 1990-12-04 Vipont Pharmaceutical, Inc. Muscosal delivery systems for treatment of periodontal disease
DE68925966T2 (en) 1988-12-22 1996-08-29 Kirin Amgen Inc CHEMICALLY MODIFIED GRANULOCYTE COLONY EXCITING FACTOR
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5198346A (en) 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
DK105489D0 (en) * 1989-03-03 1989-03-03 Novo Nordisk As POLYPEPTIDE
US5096815A (en) 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
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
US5693622A (en) 1989-03-21 1997-12-02 Vical Incorporated Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
ES2200016T3 (en) 1989-03-21 2004-03-01 Vical Incorporated EXPRESSION OF EXECUTIVE POLINUCLEOTIDIC SEQUENCES IN A VERTEBRATE.
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
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
DE59000270D1 (en) * 1989-04-11 1992-10-01 Boehringer Ingelheim Int USE OF AT LEAST ONE CYTOKIN FOR THE PRODUCTION OF A MEDICINAL PRODUCT 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
EP0394827A1 (en) 1989-04-26 1990-10-31 F. Hoffmann-La Roche Ag Chimaeric CD4-immunoglobulin 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
CA2033176C (en) * 1989-06-09 1999-12-14 Julian Richard Este Wells Growth hormone fusion proteins
JPH0327320A (en) 1989-06-26 1991-02-05 Ajinomoto Co Inc Human b cell differentiation factor pharmaceutical composition
DK0479909T3 (en) 1989-06-29 1997-04-07 Medarex Inc Bispecific reagents for AIDS treatment
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
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
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
GB8927480D0 (en) 1989-12-05 1990-02-07 Delta Biotechnology Ltd Mutant fungal strain detection and new promoter
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
IE66494B1 (en) 1989-09-26 1996-01-10 Immunex Corp Granulocyte-colony stimulating factor receptors
ATE168416T1 (en) 1989-10-05 1998-08-15 Optein Inc CELL-FREE SYNTHESIS AND ISOLATION OF GENES AND POLYPEPTIDES
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.
GB8924021D0 (en) 1989-10-25 1989-12-13 Celltech Ltd Recombinant dna method and vectors for the use therein
DK0452457T3 (en) 1989-11-03 1998-03-02 Univ Vanderbilt Method for in vivo removal of functional foreign genes
US5676954A (en) 1989-11-03 1997-10-14 Vanderbilt University Method of in vivo delivery of functioning foreign genes
US5173408A (en) 1989-11-13 1992-12-22 Lange Louis George Iii Mammalian pancreatic cholesterol esterase
US5580560A (en) * 1989-11-13 1996-12-03 Novo Nordisk A/S Modified factor VII/VIIa
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
AU7247191A (en) 1990-01-11 1991-08-05 Molecular Affinities Corporation Production of antibodies using gene libraries
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
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP1690935A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
DE69117545T2 (en) * 1990-01-25 1996-10-31 Univ Washington Factor X-LACI hybrid protein
JPH04211375A (en) 1990-02-05 1992-08-03 Ajinomoto Co Inc Synthetic gene and production of human serum albumin using the synthetic gene
US5747334A (en) 1990-02-15 1998-05-05 The University Of North Carolina At Chapel Hill Random peptide library
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
WO1991018088A1 (en) 1990-05-23 1991-11-28 The United States Of America, Represented By The Secretary, United States Department Of Commerce Adeno-associated virus (aav)-based eucaryotic vectors
US5766897A (en) * 1990-06-21 1998-06-16 Incyte Pharmaceuticals, Inc. Cysteine-pegylated proteins
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
ATE104348T1 (en) 1990-07-10 1994-04-15 Boehringer Ingelheim Int O-GLYCOSYLATED IFN-ALPHA.
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
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
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5391183A (en) * 1990-09-21 1995-02-21 Datascope Investment Corp Device and method sealing puncture wounds
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
EP0557300B1 (en) 1990-10-29 1997-11-19 Chiron Corporation Bispecific antibodies, method of production, and uses thereof
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.
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
JPH0638771A (en) 1990-10-31 1994-02-15 Tonen Corp Expression of human protein disulfide isomerase gene and production of polypeptide by co-expression with the gene
US5353535A (en) 1990-11-05 1994-10-11 Plumly George W Floor type advertising apparatus
JP3366947B2 (en) 1990-11-29 2003-01-14 アンスティテュ ナシオナル ド ラ ルシェルシュ アグロノミクーイ.エン.エル.アー. Novel mutants derived from type 1 interferon, their production method and their application
EP0564531B1 (en) 1990-12-03 1998-03-25 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
US5272080A (en) * 1991-02-19 1993-12-21 Pharmavene, Inc. Production of butyrylcholinesterase
US5833982A (en) 1991-02-28 1998-11-10 Zymogenetics, Inc. Modified factor VII
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
CA2062659A1 (en) 1991-03-12 1992-09-13 Yasutaka Igari Composition for sustained-release of erythropoietin
US5817471A (en) 1991-03-14 1998-10-06 The United States Of America As Represented By The Department Of Health And Human Services Trk tyrosine kinase receptor is the physiological receptor for nerve growth factor
DE69229454T2 (en) 1991-03-28 2000-01-05 Merck & Co Inc Subunit-C of the vascular endothelial cell growth factor
US5374617A (en) 1992-05-13 1994-12-20 Oklahoma Medical Research Foundation Treatment of bleeding with modified tissue factor in combination with FVIIa
DE69233367T2 (en) 1991-04-10 2005-05-25 The Scripps Research Institute, La Jolla LIBRARIES OF HETERODIMERIC RECEPTORS BY PHAGEMIDES
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
US5330901A (en) 1991-04-26 1994-07-19 Research Corporation Technologies, Inc. Expression of human serum albumin in Pichia pastoris
IE921342A1 (en) 1991-04-26 1992-11-04 Surface Active Ltd Novel antibodies, and methods for their use
FR2676070B1 (en) 1991-04-30 1994-09-30 Rhone Poulenc Rorer Sa YEAST PROMOTER AND ITS USE.
US5646012A (en) * 1991-04-30 1997-07-08 Rhone-Poulenc Rorer S.A. Yeast promoter and use thereof
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
AU2238292A (en) 1991-06-14 1993-01-12 Xoma Corporation 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.
US5851795A (en) * 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
US5844095A (en) * 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
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
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
US6270989B1 (en) 1991-11-05 2001-08-07 Transkaryotic Therapies, Inc. Protein production and delivery
US5641670A (en) 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
PT101031B (en) * 1991-11-05 2002-07-31 Transkaryotic Therapies Inc PROCESS FOR THE SUPPLY OF PROTEINS BY GENETIC THERAPY
DE69226197T2 (en) * 1991-11-08 1999-02-11 Somatogen Inc HEMOGLOBINE AS A MEDICINE DELIVERY SYSTEM
US5786883A (en) * 1991-11-12 1998-07-28 Pilkington Barnes Hind, Inc. Annular mask contact lenses
ATE275198T1 (en) 1991-12-02 2004-09-15 Medical Res Council PRODUCTION OF ANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES 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
US5540923A (en) * 1991-12-06 1996-07-30 Landsforeningen Til Kraeftens Bekaemplse Interferon proteins
US5428139A (en) 1991-12-10 1995-06-27 The Dow Chemical Company Bicyclopolyazamacrocyclophosphonic acid complexes for use as radiopharmaceuticals
WO1993014200A1 (en) 1992-01-07 1993-07-22 Tsi Corporation 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.
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.
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2686901A1 (en) 1992-01-31 1993-08-06 Rhone Poulenc Rorer Sa NOVEL ANTITHROMBOTIC POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US6004554A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for targeting the vasculature of solid tumors
US5230886A (en) 1992-03-18 1993-07-27 Trustees Of Boston University Tumor cell suppression
DK36392D0 (en) * 1992-03-19 1992-03-19 Novo Nordisk As USE OF CHEMICAL COMPOUND
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
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
ES2136664T3 (en) 1992-06-09 1999-12-01 Hoppe Ag CLOSING SYSTEM AND LOCKING ASSEMBLY.
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
JP3269504B2 (en) 1992-07-08 2002-03-25 三菱ウェルファーマ株式会社 Method for producing human serum albumin
FR2694294B1 (en) * 1992-07-30 1994-09-09 Rhone Poulenc Rorer Sa Yeast promoter and its user.
CA2139358C (en) 1992-07-31 2001-02-13 Barbara H. O'connor Human growth hormone aqueous formulation
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)
WO1994008599A1 (en) * 1992-10-14 1994-04-28 The Regents Of The University Of Colorado 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
FR2701953B1 (en) 1993-02-22 1995-05-24 Centre Nat Rech Scient Multi-VIP fusion protein and method for preparing recombinant VIP.
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
US5869445A (en) * 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
CA2162726A1 (en) * 1993-05-21 1994-12-08 Kathleen L. Berkner Modified factor vii
DE69434447T2 (en) 1993-06-07 2006-05-18 Vical, Inc., San Diego PLASMIDE FOR GENE THERAPY
US5621039A (en) * 1993-06-08 1997-04-15 Hallahan; Terrence W. Factor IX- polymeric conjugates
DK82893D0 (en) * 1993-07-08 1993-07-08 Novo Nordisk As PEPTIDE
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
WO1995015982A2 (en) 1993-12-08 1995-06-15 Genzyme Corporation Process for generating specific antibodies
PT1231268E (en) 1994-01-31 2005-11-30 Univ Boston BANKS OF POLYCLONE ANTIBODIES
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
US5932780A (en) 1994-02-28 1999-08-03 Yissum Research Development Company Of Hebrew University Of Jerusalem Transgenic non-human animal assay system for anti-cholinesterase substances
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
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.
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
JPH0851982A (en) 1994-08-11 1996-02-27 Asahi Glass Co Ltd Modified gene coding human serum albumen
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
AU708972B2 (en) 1994-11-07 1999-08-19 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
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
FR2726576B1 (en) 1994-11-07 1997-01-31 Pf Medicament PRODUCTION OF HYDROPHOBIC PEPTIDE-LIKE PEPTIDES, RECOMBINANT PEPTIDE, CORRESPONDING DNA SEQUENCE
AT403167B (en) 1994-11-14 1997-11-25 Immuno Ag SELECTION AND EXPRESSION OF FOREIGN PROTEINS BY MEANS OF A SELECTION-AMPLIFICATION SYSTEM
US5695750A (en) 1994-11-25 1997-12-09 The United States Of America As Represented By The Secretary Of The Army Compositions for use to deactivate organophosphates
WO1996018412A1 (en) * 1994-12-12 1996-06-20 Beth Israel Hospital Association Chimeric cytokines and uses thereof
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
US5837281A (en) * 1995-03-17 1998-11-17 Takeda Chemical Industries, Ltd. Stabilized interface for iontophoresis
TW426523B (en) 1995-04-06 2001-03-21 Hoffmann La Roche Interferon solution
EP0822984A4 (en) 1995-04-27 2000-05-03 Human Genome Sciences Inc Human tumor necrosis factor receptors
EP1709970A1 (en) 1995-04-27 2006-10-11 Abgenix, Inc. Human antibodies against EGFR, derived from immunized xenomice
CA2219486A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5480640A (en) 1995-05-02 1996-01-02 Schering Corporation Alpha interferon for treating prostate cancer
CN1515672A (en) 1995-05-05 2004-07-28 人类基因组科学公司 Human chemotactic factor beta-8, chemotactic factor beta-1 and macrophage inflammatory protein-4
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
US5705151A (en) 1995-05-18 1998-01-06 National Jewish Center For Immunology & Respiratory Medicine Gene therapy for T cell regulation
US6001625A (en) 1995-05-19 1999-12-14 The United States Of America As Represented By The Secretary Of The Army Site-directed mutagenesis of esterases
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
US5766620A (en) * 1995-10-23 1998-06-16 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
DE69632062T2 (en) 1995-11-02 2004-11-18 Schering Corp. CONTINUOUS, LOW-DOSE CYTOKINE INFUSION THERAPY
US6048964A (en) * 1995-12-12 2000-04-11 Stryker Corporation Compositions and therapeutic methods using morphogenic proteins and stimulatory factors
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
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
US6045788A (en) * 1996-02-28 2000-04-04 Cornell Research Foundation, Inc. Method of stimulation of immune response with low doses of IL-2
DE19639601A1 (en) 1996-02-28 1997-09-04 Bayer Ag Parapox viruses that contain foreign DNA, their production and their use in vaccines
US5912229A (en) * 1996-03-01 1999-06-15 Novo Nordisk Als Use of a pharmaceutical composition comprising an appetite-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
AU5711196A (en) 1996-03-14 1997-10-01 Human Genome Sciences, Inc. Apoptosis inducing molecule i
JP2000506865A (en) 1996-03-14 2000-06-06 ジ イミューン リスポンス コーポレイション Targeted delivery of genes encoding interferons
CA2248136A1 (en) * 1996-03-21 1997-09-25 Human Genome Sciences, Inc. Human endometrial specific steroid-binding factor i, ii and iii
KR20030096450A (en) 1996-03-22 2003-12-31 휴먼 게놈 사이언시즈, 인코포레이티드 Apoptosis inducing molecule ii
US6204022B1 (en) * 1996-04-12 2001-03-20 Pepgen Corporation And University Of Florida Low-toxicity human interferon-alpha analogs
AU2405297A (en) * 1996-04-23 1997-11-12 Chugai Seiyaku Kabushiki Kaisha Cerebral stroke/cerebral edema preventive or remedy containing il-8 binding inhibitor as active ingredient
WO1997044026A1 (en) * 1996-05-22 1997-11-27 Neuromedica, Inc. Compositions comprising conjugates of cis-docosahexaenoic acid and taxotere
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
AU710069B2 (en) * 1996-06-11 1999-09-16 Roche Diagnostics Gmbh Recombinant blood-coagulation proteases
ES2242227T5 (en) 1996-07-15 2011-12-09 Chugai Seiyaku Kabushiki Kaisha NEW VEGF TYPE FACTOR.
ES2316153T3 (en) 1996-08-16 2009-04-01 Human Genome Sciences, Inc. HUMAN ALFA ENDOQUINA.
AU4055697A (en) 1996-08-16 1998-03-06 Schering Corporation Mammalian cell surface antigens; related reagents
DE69739469D1 (en) 1996-08-23 2009-07-30 Vegenics Ltd Recombinant vascular endothelial growth factor D (VEGF-D)
WO1998012344A1 (en) * 1996-09-18 1998-03-26 Human Genome Sciences, Inc. Human tumor necrosis factor receptor-like genes
US6225290B1 (en) 1996-09-19 2001-05-01 The Regents Of The University Of California Systemic gene therapy by intestinal cell transformation
US5994112A (en) 1996-10-09 1999-11-30 Incyte Pharmaceuticals, Inc. Human protein tyrosine kinase
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
AU4988697A (en) 1996-10-24 1998-05-15 Vion Pharmaceuticals, Inc. Monophosphate prodrugs of beta-l-fd4c and beta-l-fddc as potent antiviral agents
KR20050004269A (en) 1996-10-25 2005-01-12 휴먼 게놈 사이언시즈, 인코포레이티드 Neutrokine alpha
JPH10134761A (en) 1996-10-30 1998-05-22 Ebara Corp Ion implantation device and method
US5908830A (en) * 1996-10-31 1999-06-01 Merck & Co., Inc. Combination therapy for the treatment of diabetes and 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
GB9623205D0 (en) * 1996-11-07 1997-01-08 Eurand Int Novel pharmaceutical compositions
CA2722378C (en) 1996-12-03 2015-02-03 Amgen Fremont Inc. Human antibodies that bind tnf.alpha.
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
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
DE69837806T3 (en) 1997-01-28 2012-01-05 Human Genome Sciences, Inc. "DEATH-DOMAIN" -INTERDENTING RECEPTOR 4 (DR4), A MEMBER OF THE TNF-RECEPTOR SUPERFAMILY, BINDING ON TRAIL (APO-2L)
WO1998032466A1 (en) 1997-01-29 1998-07-30 Polymasc Pharmaceuticals Plc Pegylation process
GB2324529A (en) 1997-02-21 1998-10-28 Merck & Co Inc A combinatorial library based on a tetrapeptide substituted with aminomethylcoumarin for characterizing proteases
US7026447B2 (en) * 1997-10-09 2006-04-11 Human Genome Sciences, Inc. 53 human secreted proteins
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds and uses thereof
BRPI9809391B8 (en) 1997-04-14 2021-05-25 Amgen Res Munich Gmbh process for producing an anti-human antigen receptor, human antibody and pharmaceutical composition
MY118835A (en) * 1997-04-18 2005-01-31 Ipsen Pharma Biotech Sustained release compositions and the process for their preparation
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998050347A1 (en) 1997-05-05 1998-11-12 The Regents Of The University Of California Naphthols useful in antiviral methods
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
JP2002503963A (en) 1997-06-11 2002-02-05 ヒューマン・ジェノム・サイエンシズ・インコーポレイテッド Human tumor necrosis factor receptor TR9
GB9713412D0 (en) 1997-06-26 1997-08-27 Delta Biotechnology Ltd Improved protein expression strains
US5858719A (en) 1997-07-17 1999-01-12 Incyte Pharmaceuticals, Inc. Polynucleotides encoding human ATP binding-cassette transport protein and methods of use
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
ES2186660T3 (en) 1997-09-21 2003-05-16 Schering Corp COMBINATION THERAPY TO ERADICATE HCV-RNA DETECTABLE IN PATIENTS WITH CHRONIC HEPATITIS C INFECTION.
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
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
WO1999029314A1 (en) * 1997-12-08 1999-06-17 Bristol-Myers Squibb Company Novel salts of metformin and method
EP1037927B1 (en) * 1997-12-08 2004-05-19 Lexigen Pharmaceuticals Corp. Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
US6221378B1 (en) * 1998-02-10 2001-04-24 Generex Pharmaceuticals Incorporated Mixed micellar delivery system and method of preparation
US6017545A (en) * 1998-02-10 2000-01-25 Modi; Pankaj Mixed micellar delivery system and method of preparation
DE19813802A1 (en) 1998-03-27 1999-11-11 Retro Tech Gmbh Anti-viral effects of propolis through inhibition of viral nucleic acid polymerases
GB9806631D0 (en) 1998-03-28 1998-05-27 Safeglass Europ Limited Safetyglass
US6251868B1 (en) 1998-04-30 2001-06-26 Teijin Limited Method for treating a human immunodeficiency virus infection
WO1999059621A1 (en) * 1998-05-15 1999-11-25 Schering Corporation Combination therapy comprising ribavirin and interferon alpha in antiviral treatment naive patients having g chronic hepatitis c 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
US5970300A (en) * 1998-06-01 1999-10-19 Xerox Corporation Apparatus for applying scents to paper in a printer/copier
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
MXPA00012842A (en) 1998-06-24 2004-06-22 Univ Emory Use of 3'-azido-2',3'-dideoxyuridine in combination with further anti-hiv drugs for the manufacture of a medicament for the treatment of hiv.
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
US6346543B1 (en) * 1998-08-17 2002-02-12 Aventis Pharma S.A. Use of a taxoid to treat abnormal cell proliferation in the brain
US6193997B1 (en) * 1998-09-27 2001-02-27 Generex Pharmaceuticals Inc. Proteinic drug delivery system using membrane mimetics
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
US20030190669A1 (en) * 1998-12-30 2003-10-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
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-!}
PT1187852E (en) 1999-05-19 2007-11-14 Merck Patent Gmbh Expression and export of interferon-alpha proteins as fc fusion proteins
JO2291B1 (en) * 1999-07-02 2005-09-12 اف . هوفمان لاروش ايه جي Erythopintin derivatives
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
AU1573601A (en) 1999-10-21 2001-04-30 Monsanto Company Post-translational modification of recombinant proteins produced in plants
US6569832B1 (en) * 1999-11-12 2003-05-27 Novo Nordisk A/S Inhibition of beta cell degeneration
CA2747325A1 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20050100991A1 (en) 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
DK1311285T4 (en) * 2000-05-15 2017-07-24 Hoffmann La Roche Liquid pharmaceutical composition containing an erythropoietin derivative
US7101561B2 (en) * 2000-12-01 2006-09-05 Innogenetics N.V. Purified hepatitis C virus envelope proteins for diagnostic and therapeutic use
BR0116024A (en) 2000-12-07 2005-12-13 Lilly Co Eli Heterologous Fusion Protein and Use thereof
US7070973B2 (en) 2000-12-26 2006-07-04 Board Of Regents Of The University Of Nebraska Butyrylcholinesterase variants and methods of use
US20050054051A1 (en) 2001-04-12 2005-03-10 Human Genome Sciences, Inc. Albumin fusion proteins
US20060084794A1 (en) * 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
US20030143191A1 (en) * 2001-05-25 2003-07-31 Adam Bell Chemokine beta-1 fusion proteins
WO2003013573A1 (en) 2001-08-10 2003-02-20 Epix Medical, Inc. Polypeptide conjugates with extended circulating half-lives
AU2002332041A1 (en) 2001-10-05 2003-04-22 Human Genome Sciences, Inc. Albumin fusion proteins
ES2606840T3 (en) 2001-10-10 2017-03-28 Ratiopharm Gmbh Remodeling and glycoconjugation of granulocyte colony stimulating factor (G-CSF)
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
WO2005003296A2 (en) 2003-01-22 2005-01-13 Human Genome Sciences, Inc. Albumin fusion proteins
AU2002353374A1 (en) 2001-12-21 2003-07-09 Nexia Biotechnologies, Inc. Production of butyrylcholinesterases in transgenic mammals
EP1463752A4 (en) * 2001-12-21 2005-07-13 Human Genome Sciences Inc Albumin fusion proteins
US20080167238A1 (en) 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
EP1463751B1 (en) 2001-12-21 2013-05-22 Human Genome Sciences, Inc. Albumin fusion proteins
US20080194481A1 (en) 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
MXPA04012496A (en) * 2002-06-21 2005-09-12 Novo Nordisk Healthcare Ag Pegylated factor vii glycoforms.
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
US6913890B2 (en) * 2002-12-18 2005-07-05 Palo Alto Research Center Incorporated Process for preparing albumin protein conjugated oligonucleotide probes
ES2453105T3 (en) * 2003-02-27 2014-04-04 Baxter International Inc. Device for calibration in a method for certifiable inactivation of pathogens by irradiation in a biological fluid
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
US7436410B2 (en) * 2005-04-01 2008-10-14 Seiko Epson Corporation System and method for programming a controller
AU2006280312A1 (en) 2005-08-12 2007-02-22 Human Genome Sciences, Inc. Albumin fusion proteins
US7438904B1 (en) 2005-10-04 2008-10-21 University Of Kentucky Research Foundation High-activity mutants of butyrylcholinesterase for cocaine hydrolysis and method of generating the same
CA2562249A1 (en) 2005-10-20 2007-04-20 University Of Ottawa Heart Institute Anf analogue
EP1816201A1 (en) * 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
AU2007258609B2 (en) 2006-06-07 2013-01-24 Human Genome Sciences, Inc. Albumin fusion proteins
SI2717898T1 (en) * 2011-06-10 2019-07-31 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof

Also Published As

Publication number Publication date
AU2001261024A1 (en) 2001-10-30
AU2001259066A1 (en) 2001-10-30
WO2001079258A9 (en) 2002-02-28
US20080261877A1 (en) 2008-10-23
US20050266532A1 (en) 2005-12-01
US20080131399A1 (en) 2008-06-05
ES2529300T3 (en) 2015-02-18
EP1276756A4 (en) 2004-06-09
EP2213743A1 (en) 2010-08-04
EP1274719A4 (en) 2004-05-19
CA2405557A1 (en) 2001-10-25
EP1983055A1 (en) 2008-10-22
DK2236152T3 (en) 2014-07-07
US6994857B2 (en) 2006-02-07
US20140010798A1 (en) 2014-01-09
CA2405701A1 (en) 2001-10-25
WO2001079443A3 (en) 2002-02-21
WO2001079444A2 (en) 2001-10-25
CA2405563A1 (en) 2001-10-25
US10080785B2 (en) 2018-09-25
US7482013B2 (en) 2009-01-27
CA2405557C (en) 2013-09-24
FR16C0043I2 (en) 2020-04-10
US20180200346A1 (en) 2018-07-19
US20080269126A1 (en) 2008-10-30
CA2405709A1 (en) 2001-10-25
JP2014057589A (en) 2014-04-03
EP2275557A1 (en) 2011-01-19
US7785599B2 (en) 2010-08-31
JP2003530847A (en) 2003-10-21
JP2003530839A (en) 2003-10-21
WO2001079480A1 (en) 2001-10-25
WO2001079442A2 (en) 2001-10-25
US9821039B2 (en) 2017-11-21
EP2311872A1 (en) 2011-04-20
EP2206720A1 (en) 2010-07-14
EP1274720A1 (en) 2003-01-15
US20080269125A1 (en) 2008-10-30
US20030219875A1 (en) 2003-11-27
EP1832599A3 (en) 2007-11-21
AU2001262942A1 (en) 2001-10-30
EP1276856A4 (en) 2004-06-09
US9849162B2 (en) 2017-12-26
EP1276849A4 (en) 2004-06-09
WO2001079442A3 (en) 2002-06-06
WO2001077137A1 (en) 2001-10-18
AU2001266557A1 (en) 2001-10-23
WO2001079444A3 (en) 2002-05-23
AU2001259063A1 (en) 2001-10-30
CA2405525A1 (en) 2001-10-25
WO2001079271A9 (en) 2002-02-28
US20040171123A1 (en) 2004-09-02
US20090285816A9 (en) 2009-11-19
EP2267026A1 (en) 2010-12-29
US20080267962A1 (en) 2008-10-30
JP2003530838A (en) 2003-10-21
EP1278544A2 (en) 2003-01-29
US20070287173A9 (en) 2007-12-13
EP1276756A1 (en) 2003-01-22
US20080269127A1 (en) 2008-10-30
WO2001077137A9 (en) 2002-05-02
CA2747325A1 (en) 2001-10-25
US20120141449A1 (en) 2012-06-07
US9775888B2 (en) 2017-10-03
JP2003531590A (en) 2003-10-28
US20140004095A1 (en) 2014-01-02
US6905688B2 (en) 2005-06-14
JP2011217750A (en) 2011-11-04
US20050266533A1 (en) 2005-12-01
US20030199043A1 (en) 2003-10-23
EP2216409B1 (en) 2014-12-03
EP2236152A1 (en) 2010-10-06
US7507414B2 (en) 2009-03-24
US20030125247A1 (en) 2003-07-03
AU2001264563A1 (en) 2001-10-30
CA2405912A1 (en) 2001-10-18
US8946156B2 (en) 2015-02-03
US20080269128A1 (en) 2008-10-30
ES2484966T3 (en) 2014-08-12
US20120252732A1 (en) 2012-10-04
DK2216409T3 (en) 2015-01-05
EP1278767A1 (en) 2003-01-29
EP2298355A3 (en) 2011-06-29
EP2067488A1 (en) 2009-06-10
EP1278767A4 (en) 2003-11-12
US6926898B2 (en) 2005-08-09
EP1276849A2 (en) 2003-01-22
EP1274719A2 (en) 2003-01-15
EP1278544A4 (en) 2004-08-18
EP1832599A2 (en) 2007-09-12
US20040010134A1 (en) 2004-01-15
WO2001079271A1 (en) 2001-10-25
EP2216409A1 (en) 2010-08-11
EP2357008A1 (en) 2011-08-17
US20110280830A9 (en) 2011-11-17
EP2298355A2 (en) 2011-03-23
US20120141415A1 (en) 2012-06-07
FR16C0043I1 (en) 2016-12-09
WO2001079443A2 (en) 2001-10-25
BE2016C059I2 (en) 2020-08-20
EP1274720A4 (en) 2004-08-18
US20100189686A1 (en) 2010-07-29
JP2004506407A (en) 2004-03-04
EP1803730A1 (en) 2007-07-04
US20130266553A1 (en) 2013-10-10
JP2003530852A (en) 2003-10-21
AU2001274809A1 (en) 2001-10-30
US20030171267A1 (en) 2003-09-11
JP2003530846A (en) 2003-10-21
EP1276856A1 (en) 2003-01-22
EP2236152B1 (en) 2014-06-04
WO2001079258A1 (en) 2001-10-25
EP2295456A1 (en) 2011-03-16
WO2001079480A9 (en) 2003-09-18

Similar Documents

Publication Publication Date Title
CA2513213C (en) Albumin fusion proteins
CA2405550A1 (en) Albumin fusion proteins
AU2008319332B2 (en) Albumin fusion proteins
US20060084794A1 (en) Albumin fusion proteins
CA2654055A1 (en) Albumin fusion proteins
US20120021985A1 (en) Albumin fusion proteins
CA2554089A1 (en) Albumin fusion proteins
CA2618476A1 (en) Albumin fusion proteins
CA2663352A1 (en) Albumin fusion proteins
CA2446739A1 (en) Chemokine beta-1 fusion proteins
US20070253973A1 (en) Fusion proteins comprising alpha fetoprotein

Legal Events

Date Code Title Description
FZDE Discontinued