US20100291680A1 - Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission - Google Patents

Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission Download PDF

Info

Publication number
US20100291680A1
US20100291680A1 US12/791,983 US79198310A US2010291680A1 US 20100291680 A1 US20100291680 A1 US 20100291680A1 US 79198310 A US79198310 A US 79198310A US 2010291680 A1 US2010291680 A1 US 2010291680A1
Authority
US
United States
Prior art keywords
peptides
peptide
seq
hiv
virus
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
US12/791,983
Inventor
Dani Paul Bolognesi
Thomas James Matthews
Carl T. Wild
Shawn O'Lin Barney
Dennis Michael Lambert
Stephen Robert Petteway, Jr.
Alphonse J. Langlois
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.)
Trimeris Inc
Original Assignee
Trimeris Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27000760&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100291680(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US08/255,208 external-priority patent/US6440656B1/en
Priority claimed from US08/360,107 external-priority patent/US6017536A/en
Application filed by Trimeris Inc filed Critical Trimeris Inc
Priority to US12/791,983 priority Critical patent/US20100291680A1/en
Publication of US20100291680A1 publication Critical patent/US20100291680A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • 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
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/21Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/22Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Neisseriaceae (F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/245Escherichia (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/285Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pasteurellaceae (F), e.g. Haemophilus influenza
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/305Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Micrococcaceae (F)
    • C07K14/31Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Micrococcaceae (F) from Staphylococcus (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0806Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0812Tripeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0815Tripeptides with the first amino acid being basic
    • C07K5/0817Tripeptides with the first amino acid being basic the first amino acid being Arg
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0819Tripeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1008Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/101Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1013Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1016Tetrapeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1021Tetrapeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1024Tetrapeptides with the first amino acid being heterocyclic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16211Lymphocryptovirus, e.g. human herpesvirus 4, Epstein-Barr Virus
    • C12N2710/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/14011Deltaretrovirus, e.g. bovine leukeamia virus
    • C12N2740/14022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/17011Spumavirus, e.g. chimpanzee foamy virus
    • C12N2740/17022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18411Morbillivirus, e.g. Measles virus, canine distemper
    • C12N2760/18422New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18611Respirovirus, e.g. Bovine, human parainfluenza 1,3
    • C12N2760/18622New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18711Rubulavirus, e.g. mumps virus, parainfluenza 2,4
    • C12N2760/18722New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/82Proteins from microorganisms
    • Y10S530/826Viruses

Definitions

  • the present invention relates, first, to DP178 (SEQ ID NO:1), a peptide corresponding to amino acids 638 to 673 of the HIV-1 LAI transmembrane protein (TM) gp41, and portions or analogs of DP178 (SEQ ID NO:1), which exhibit anti-membrane fusion capability, antiviral activity, such as the ability to inhibit HIV transmission to uninfected CD-4 + cells, or an ability to modulate intracellular processes involving coiled-coil peptide structures. Further, the invention relates to the use of DP178 (SEQ ID NO:1) and DP178 portions and/or analogs as antifusogenic or antiviral compounds or as inhibitors of intracellular events involving coiled-coil peptide structures.
  • the present invention also relates to peptides analogous to DP107, a peptide corresponding to amino acids 558 to 595 of the HIV-1 LAI transmembrane protein (TM) gp41, having amino acid sequences present in other viruses, such as enveloped viruses, and/or other organisms, and further relates to the uses of such peptides. These peptides exhibit anti-membrane fusion capability, antiviral activity, or the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • the present invention additionally relates to methods for identifying compounds that disrupt the interaction between DP178 and DP107, and/or between DP107-like and DP178-like peptides.
  • Membrane fusion is a ubiquitous cell biological process (for a review, see White, J. M., 1992, Science 258:917-924). Fusion events which mediate cellular housekeeping functions, such as endocytosis, constitutive secretion, and recycling of membrane components, occur continuously in all eukaryotic cells.
  • fusion events occur in specialized cells. Intracellularly, for example, fusion events are involved in such processes as occur in regulated exocytosis of hormones, enzymes and neurotransmitters. Intercellularly, such fusion events feature prominently in, for example, sperm-egg fusion and myoblast fusion.
  • Fusion events are also associated with disease states. For example, fusion events are involved in the formation of giant cells during inflammatory reactions, the entry of all enveloped viruses into cells, and, in the case of human immunodeficiency virus (HIV), for example, are responsible for the virally induced cell-cell fusion which leads to cell death.
  • HIV human immunodeficiency virus
  • HIV human immunodeficiency virus
  • AIDS acquired immune deficiency syndrome
  • Retroviruses are small enveloped viruses that contain a diploid, single-stranded RNA genome, and replicate via a DNA intermediate produced by a virally-encoded reverse transcriptase, an RNA-dependent DNA polymerase (Varmus, H., 1988, Science 240:1427-1439).
  • retroviruses include, for example, oncogenic viruses such as human T-cell leukemia viruses (HTLV-I, -II, -III), and feline leukemia virus.
  • the HIV viral particle consists of a viral core, composed of capsid proteins, that contains the viral RNA genome and those enzymes required for early replicative events.
  • Myristylated Gag protein forms an outer viral shell around the viral core, which is, in turn, surrounded by a lipid membrane enveloped derived from the infected cell membrane.
  • the HIV enveloped surface glycoproteins are synthesized as a single 160 Kd precursor protein which is cleaved by a cellular protease during viral budding into two glycoproteins, gp41 and gp120.
  • gp41 is a transmembrane protein and gp120 is an extracellular protein which remains non-covalently associated with gp41, possibly in a trimeric or multimeric form (Hammarskjold, M. and Rekosh, D., 1989, Biochem. Biophys. Acta 989:269-280).
  • HIV is targeted to CD-4 + cells because the CD-4 cell surface protein acts as the cellular receptor for the HIV-1 virus (Dalgleish, A. et al., 1984, Nature 312:763-767; Klatzmann et al., 1984, Nature 312:767-768; Maddon et al., 1986, Cell 47:333-348).
  • Viral entry into cells is dependent upon gp120 binding the cellular CD-4 + receptor molecules (McDougal, J. S. et al., 1986, Science 231:382-385; Maddon, P. J. et al., 1986, Cell 47:333-348) and thus explains HIV's tropism for CD-4 + cells, while gp41 anchors the enveloped glycoprotein complex in the viral membrane.
  • HIV infection is pandemic and HIV associated diseases represent a major world health problem.
  • considerable effort is being put into the successful design of effective therapeutics, currently no curative anti-retroviral drugs against AIDS exist.
  • several stages of the HIV life cycle have been considered as targets for therapeutic intervention (Mitsuya, H. et al., 1991, FASEB J. 5:2369-2381).
  • virally encoded reverse transcriptase has been one focus of drug development.
  • a number of reverse-transcriptase-targeted drugs including 2′,3′-dideoxynucleoside analogs such as AZT, ddI, ddC, and d4T have been developed which have been shown to been active against HIV (Mitsuya. H. et al. 1991. Science 249:1533-1544). While beneficial, these nucleoside analogs are not curative, probably due to the rapid appearance of drug resistant HIV mutants (Lander, B. et al., 1989, Science 243:1731-1734). In addition, the drugs often exhibit toxic side effects such as bone marrow suppression, vomiting, and liver function abnormalities.
  • Late stage processing is dependent on the activity of a viral protease, and drugs are being developed which inhibit this protease (Erickson, J., 1990, Science 249:527-533). The clinical outcome of these candidate drugs is still in question.
  • HIV-1 enveloped proteins (gp160, gp120, gp41) have been shown to be the major antigens for anti-HIV antibodies present in AIDS patients (Barin, et al., 1985, Science 228:1094-1096). Thus far, therefore, these proteins seem to be the most promising candidates to act as antigens for anti-HIV vaccine development.
  • several groups have begun to use various portions of gp160, gp120, and/or gp41 as immunogenic targets for the host immune system. See for example, Ivanoff, L. et al., U.S. Pat. No. 5,141,867; Saith, G.
  • the present invention relates, first, to DP178 (SEQ ID NO:1), a 36-amino acid synthetic peptide corresponding to amino acids 638 to 673 of the transmembrane protein (TM) gp41 from the HIV-1 isolate LAI (HIV-1 LAI ), which exhibits potent anti-HIV-1 activity.
  • DP178 SEQ ID NO:1
  • TM transmembrane protein
  • the DP178 (SEQ ID NO:1) antiviral activity is so high that, on a weight basis, no other known anti-HIV agent is effective at concentrations as low as those at which DP178 (SEQ ID NO:1) exhibits its inhibitory effects.
  • the invention further relates to those portions and analogs of DP178 which also show such antiviral activity, and/or show anti-membrane fusion capability, or an ability to modulate intracellular processes involving coiled-coil peptide structures.
  • DP178 analog refers to a peptide which contains an amino acid sequence corresponding to the DP178 peptide sequence present within the gp41 protein of HIV-1 LAI , but found in viruses and/or organisms other than HIV-1 LAI .
  • Such DP178 analog peptides may, therefore, correspond to DP178-like amino acid sequences present in other viruses, such as, for example, enveloped viruses, such as retroviruses other than HIV-1 LAI , as well as non-enveloped viruses.
  • analogous DP178 peptides may also correspond to DP178-like amino acid sequences present in nonviral organisms.
  • DP107 is a peptide corresponding to amino acids 558-595 of the HIV-1 LAI transmembrane protein (TM) gp41.
  • TM transmembrane protein
  • DP107 analog refers to a peptide which contains an amino acid sequence corresponding to the DP107 peptide sequence present within the gp41 protein of HIV-1 LAI , but found in viruses and organisms other than HIV-1 LAI .
  • Such DP107 analog peptides may, therefore, correspond to DP107-like amino acid sequences present in other viruses, such as, for example, enveloped viruses, such as retroviruses other than HIV-1 LAI , as well as non-enveloped viruses.
  • DP107 analog peptides may also correspond to DP107-like amino acid sequences present in nonviral organisms.
  • the peptides of the invention include DP107 analog and DP178 analog peptides having amino acid sequences recognized or identified by the 107 ⁇ 178 ⁇ 4, ALLMOTI5 and/or PLZIP search motifs described herein.
  • the peptides of the invention may, for example, exhibit antifusogenic activity, antiviral activity, and/or may have the ability to modulate intracellular processes which involve coiled-coil peptide structures.
  • antiviral activity of the peptides of the invention includes, but is not limited to the inhibition of HIV transmission to uninfected CD-4 + cells.
  • the antifusogenic capability, antiviral activity or intracellular modulatory activity of the peptides of the invention merely requires the presence of the peptides of the invention, and, specifically, does not require the stimulation of a host immune response directed against such peptides.
  • the peptides of the invention may be used, for example, as inhibitors of membrane fusion-asociated events, such as, for example, the inhibition of human and non-human retroviral, especially HIV, transmission to uninfected cells. It is further contemplated that the peptides of the invention may be used as modulators of intracellular events involving coiled-coil peptide structures.
  • the peptides of the invention may, alternatively, be used to identify compounds which may themselves exhibit antifusogenic, antiviral, or intracellular modulatory activity. Additional uses include, for example, the use of the peptides of the invention as organism or viral type and/or subtype-specific diagnostic tools.
  • anti-membrane fusion refers to an agent's ability to inhibit or reduce the level of membrane fusion events between two or more moieties relative to the level of membrane fusion which occurs between said moieties in the absence of the peptide.
  • the moieties may be, for example, cell membranes or viral structures, such as viral envelopes or pili.
  • antiviral refers to the compound's ability to inhibit viral infection of cells, via, for example, cell-cell fusion or free virus infection.
  • Such infection may involve membrane fusion, as occurs in the case of enveloped viruses, or some other fusion event involving a viral structure and a cellular structure (e.g., such as the fusion of a viral pilus and bacterial membrane during bacterial conjugation). It is also contemplated that the peptides of the invention may exhibit the ability to modulate intracellular events involving coiled-coil peptide structures. “Modulate”, as used herein, refers to a stimulatory or inhibitory effect on the intracellular process of interest relative to the level or activity of such a process in the absence of a peptide of the invention.
  • Embodiments of the invention are demonstrated below wherein an extremely low concentration of DP178 (SEQ ID NO:1), and very low concentrations of a DP178 homolog (SEQ ID NO:3) are shown to be potent inhibitors of HIV-1 mediated CD-4 + cell-cell fusion (i.e., syncytial formation) and infection of CD-4 + cells by cell-free virus. Further, it is shown that DP178 (SEQ ID NO:1) is not toxic to cells, even at concentrations 3 logs higher than the inhibitory DP-178 (SEQ ID NO:1) concentration.
  • the present invention is based, in part, on the surprising discovery that the DP107 and DP178 domains of the HIV gp41 protein non-covalently complex with each other, and that their interaction is required for the normal infectivity of the virus. This discovery is described in the Example presented, below, in Section 8. The invention, therefore, further relates to methods for identifying antifusogenic, including antiviral, compounds that disrupt the interaction between DP107 and DP178, and/or between DP107-like and DP178-like peptides.
  • Peptides are defined herein as organic compounds comprising two or more amino acids covalently joined by peptide bonds. Peptides may be referred to with respect to the number of constituent amino acids, i.e., a dipeptide contains two amino acid residues, a tripeptide contains three, etc. Peptides containing ten or fewer amino acids may be referred to as oligopeptides, while those with more than ten amino acid residues are polypeptides. Such peptides may also include any of the modifications and additional amino and carboxy groups as are described herein.
  • FIG. 1 Amino acid sequence of DP178 (SEQ ID NO:1) derived from HIV LAI ; DP178 homologs derived from HIV-1 SF2 (DP-185; SEQ ID NO:3), HIV-1 RF (SEQ ID NO:4), and HIV-1 MN (SEQ ID NO:5); DP178 homologs derived from amino acid sequences of two prototypic HIV-2 isolates, namely, HIV-2 rod (SEQ ID NO:6) and HIV-2 NIHZ (SEQ ID NO:7); control peptides: DP-180 (SEQ ID NO:2), a peptide incorporating the amino acid residues of DP178 in a scrambled sequence; DP-118 (SEQ ID NO:10) unrelated to DP178, which inhibits HIV-1 cell free virus infection; DP-125 (SEQ ID NO:8), unrelated to DP178, also inhibits HIV-1 cell free virus infection; DP-116 (SEQ ID NO:9), unrelated to DP178, is negative for inhibition of
  • FIG. 2 Inhibition of HIV-1 cell-free virus infection by synthetic peptides.
  • IC 50 refers to the concentration of peptide that inhibits RT production from infected cells by 50% compared to the untreated control.
  • Control the level of RT produced by untreated cell cultures infected with the same level of virus as treated cultures.
  • FIG. 3 Inhibition of HIV-1 and HIV-2 cell-free virus infection by the synthetic peptide DP178 (SEQ ID NO:1).
  • IC 50 concentration of peptide that inhibits RT production by 50% compared to the untreated control.
  • Control Level of RT produced by untreated cell cultures infected with the same level of virus as treated cultures.
  • FIG. 4A-4B Fusion Inhibition Assays.
  • FIG. 4A DP178 (SEQ ID NO:1) inhibition of HIV-1 prototypic isolate-mediated syncytial formation; data represents the number of virus-induced syncytial per cell.
  • FIG. 4B DP-180 (SEQ ID NO:2) represents a scrambled control peptide; DP-185 (SEQ ID NO:3) represents a DP178 homolog derived from HIV-1 SF2 isolate; Control, refers to the number of syncytial produced in the absence of peptide.
  • FIG. 5 Fusion inhibition assay: HIV-1 vs. HIV-2. Data represents the number of virus-induced syncytial per well. ND: not done.
  • FIG. 6 Cytotoxicity study of DP178 (SEQ ID NO:1) and DP-116 (SEQ ID NO:9) on CEM cells. Cell proliferation data is shown.
  • FIG. 7 Schematic representation of HIV-gp41 and maltose binding protein (MBP)-gp41 fusion proteins.
  • DP107 and DP178 are synthetic peptides based on the two putative helices of gp41.
  • the letter P in the DP107 boxes denotes an Ile to Pro mutation at amino acid number 578.
  • Amino acid residues are numbered according to Meyers et al., “Human Retroviruses and AIDS”, 1991, Theoret. Biol. and Biophys. Group, Los Alamos Natl. Lab., Los Alamos, N. Mex. The proteins are more fully described, below, in Section 8.1.1.
  • FIG. 8 A point mutation alters the conformation and anti-HIV activity of M41.
  • FIG. 9 Abrogation of DP178 anti-HIV activity.
  • Cell fusion assays were carried out in the presence of 10 nM DP178 and various concentrations of M41 ⁇ 178 or M41P ⁇ 178.
  • FIG. 10 Binding of DP178 to leucine zipper of gp41 analyzed by FAb-D ELISA.
  • FIG. 11A-B Models for a structural transition in the HIV-1 TM protein. Two models are proposed which indicate a structural transition from a native oligomer to a fusogenic state following a trigger event (possibly gp120 binding to CD4).
  • FIG. 12 Motif design using heptad repeat positioning of amino acids of known coiled-coils GCN4:(SEQ ID NO:84); C-FOS:(SEQ ID NO:85); C-JUN:(SEQ ID NO:86); C-MYC:(SEQ ID NO:87); FLU LOOP 36:(SEQ ID NO:88).
  • FIG. 13 Motif design using proposed heptad repeat positioning of amino acids of DP107 and DP178.
  • FIG. 14 Hybrid motif design crossing GCN4 and DP107.
  • FIG. 15 Hybrid motif design crossing GCN4 and DP178.
  • FIG. 16 Hybrid motif design 107 ⁇ 178 ⁇ 4, crossing DP107 (SEQ ID NO:89) and DP178 (SEQ ID NO:1). This motif was found to be the most consistent at identifying relevant DP107-like and DP178-like peptide regions.
  • FIG. 17 Hybrid motif design crossing GCN4, DP107, and DP178.
  • FIG. 18 Hybrid motif design ALLMOTI5 crossing GCN4, DP107, DP178, c-Fos c-Jun, c-Myc, and Flu Loop 36.
  • FIG. 19 PLZIP motifs designed to identify N-terminal proline-leucine zipper motifs.
  • FIG. 20 Search results for HIV-1 (BRU isolate) enveloped protein gp41 (SEQ ID NO:90). Sequence search motif designations: Spades ( ): 107 ⁇ 178 ⁇ 4; Hearts ( ⁇ ) ALLMOTI5; Clubs ( ): PLZIP; Diamonds ( ⁇ ): transmembrane region (the putative transmembrane domains were identified using a PC/Gene program designed to search for such peptide regions). Asterisk (*): Lupas method. The amino acid sequences identified by each motif are bracketed by the respective characters. Representative sequences chosen based on 107 ⁇ 178 ⁇ 4 searches are underlined and in bold. DP107 and DP178 sequences are marked, and additionally double-underlined and italicized.
  • FIG. 21 Search results for human respiratory syncytial virus (RSV) strain A2 fusion glycoprotein F1 (SEQ ID NO:91). Sequence search motif designations are as in FIG. 20 .
  • FIG. 22 Search results for simian immunodeficiency virus (SIV) enveloped protein gp41 (AGM3 isolate) (SEQ ID NO:92). Sequence search motif designations are as in FIG. 20 .
  • FIG. 23 Search results for canine distemper virus (strain Onderstepoort) fusion glycoprotein 1 (SEQ ID NO:93). Sequence search motif designations are as in FIG. 20 .
  • FIG. 24 Search results for newcastle disease virus (strain Australia-Victoria/32) fusion glycoprotein F1 (SEQ ID NO:94). Sequence search motif designations are as in FIG. 20 .
  • FIG. 25 Search results for human parainfluenza 3 virus (strain NIH 47885) fusion glycoprotein F1 (SEQ ID NO:95). Sequence search motif designations are as in FIG. 20 .
  • FIG. 26 Search results for influenza A virus (strain A/AICHI/2/68) hemagglutinin precursor HA2 (SEQ ID NO:96). Sequence search designations are as in FIG. 20 .
  • FIG. 27A-F Respiratory Syncytial Virus (RSV) (SEQ ID NO:97) peptide antiviral and circular dichroism data.
  • FIG. 27A-C Peptides derived from the F2 DP178/DP107-like region T-22:(SEQ ID NO:121); T-68:(SEQ ID NO:122); T-334:(SEQ ID NO:123); T-371:(SEQ ID NO:124); T-372:(SEQ ID NO:125); T-373:(SEQ ID NO:126); T-374:(SEQ ID NO:127); T-375:(SEQ ID NO:128); T-575:(SEQ ID NO:129).
  • Antiviral and CD data FIG.
  • 27D-F Peptides derived from the F1 DP107-like region F1-107:(SEQ ID NO:98); T-12:(SEQ ID NO:130); T-13:(SEQ ID NO:131); T-15:(SEQ ID NO:132); T-19:(SEQ ID NO:133); T-28:(SEQ ID NO:134); T-30:(SEQ ID NO:135); T-66:(SEQ ID NO:136); T-576:(SEQ ID NO:137). Peptide and CD data.
  • Antiviral activity is represented by the following qualitative symbols:
  • CD data referring to the level of helicity is represented by the following qualitative symbol:
  • IC 50 refers to the concentration of peptide necessary to produce only 50% of the number of syncytial relative to infected control cultures containing no peptide. IC 50 values were obtained using purified peptides only.
  • FIG. 28A-C Respiratory Syncytial Virus (RSV) DP178-like region (F1) peptide antiviral and CD data.
  • Antiviral symbols, CD symbols, and IC 50 are as in FIG. 27A-F .
  • IC 50 values were obtained using purified peptides only.
  • FIG. 29A-E Peptides derived from the HPIV3 F1 DP107-like region. Peptide antiviral and CD data. Antiviral symbols, CD symbols, and IC 50 are as in FIG. 27A-F . Purified peptides were used to obtain IC 50 values, except where the values are marked by an asterisk (*), in which cases, the IC 50 values were obtained using a crude peptide preparation.
  • FIG. 30A-C Peptides derived from the HPIV3 F1 DP178-like region. Peptide antiviral and CD data. Antiviral symbols, CD symbols, and IC 50 are as in FIG. 27A-F . Purified peptides were used to obtain IC 50 values, except where the values are marked by an asterisk (*), in which cases, the IC 50 values were obtained using a crude peptide preparation.
  • FIG. 31 Motif search results for simian immunodeficiency virus (SIV) isolate MM251, enveloped polyprotein gp41 (SEQ ID NO:102). Sequence search designations are as in FIG. 20 .
  • FIG. 32 Motif search results for Epstein-Barr Virus (Strain B95-8), glycoprotein gp110 precursor (designated gp115) BALF4 (SEQ ID NO:103). Sequence search designations are as in FIG. 20 .
  • FIG. 33 Motif search results for Epstein-Barr Virus (Strain B95-8), BZLF1 trans-activator protein (designated EB1 or Zebra) (SEQ ID NO:104). Sequence search designations are as in FIG. 20 . Additionally, “@” refers to a well known DNA binding domain and “+” refers to a well known dimerization domain, as defined by Flemington and Speck (Flemington, E. and Speck, S. H., 1990, Proc. Natl. Acad. Sci. USA 87:9459-9463).
  • FIG. 34 Motif search results for measles virus (strain Edmonston), fusion glycoprotein F1 (SEQ ID NO:105). Sequence search designations are as in FIG. 20 .
  • FIG. 35 Motif search results for Hepatitis B Virus (Subtype AYW), major surface antigen precursor S (SEQ ID NO:106). Sequence search designations are as in FIG. 20 .
  • FIG. 36 Motif search results for simian Mason-Pfizer monkey virus, enveloped (TM) protein gp20 (SEQ ID NO:107). Sequence search designations are as in FIG. 20 .
  • FIG. 37 Motif search results for Pseudomonas aerginosa , fimbrial protein (Pilin) (SEQ ID NO:108). Sequence search designations are as in FIG. 20 .
  • FIG. 38 Motif search results for Neisseria gonorrhoeae fimbrial protein (Pilin) (SEQ ID NO:109). Sequence search designations are as in FIG. 20 .
  • FIG. 39 Motif search results for Hemophilus influenzae fimbrial protein (SEQ ID NO:110). Sequence search designations are as in FIG. 20 .
  • FIG. 40 Motif search results for Staphylococcus aureus , toxic shock syndrome toxin-1 (SEQ ID NO:111). Sequence search designations are as in FIG. 20 .
  • FIG. 41 Motif search results for Staphylococcus aureus enterotoxin Type E (SEQ ID NO:112). Sequence search designations are as in FIG. 20 .
  • FIG. 42 Motif search results for Staphylococcus aureus enterotoxin A (SEQ ID NO:113). Sequence search designations are as in FIG. 20 .
  • FIG. 43 Motif search results for Escherichia coli , heat labile enterotoxin A (SEQ ID NO:114). Sequence search designations are as in FIG. 20 .
  • FIG. 44 Motif search results for human c-fos proto-oncoprotein (SEQ ID NO:115). Sequence search designations are as in FIG. 20 .
  • FIG. 45 Motif search results for human lupus KU autoantigen protein P70 (SEQ ID NO:116). Sequence search designations are as in FIG. 20 .
  • FIG. 46 Motif search results for human zinc finger protein 10 (SEQ ID NO:117). Sequence search designations are as in FIG. 20 .
  • FIG. 47A-B Measles virus (MeV) fusion protein DP178-like region antiviral and CD data T-252AO:(SEQ ID NO:118); T-268AO:(SEQ ID NO:119).
  • Antiviral symbols, CD symbols, and IC 50 are as in FIG. 27A-F . IC 50 values were obtained using purified peptides.
  • FIG. 48A-B Simian immunodeficiency virus (SIV) TM (fusion) protein DP178-like region antiviral data (SEQ ID NO:120). Antiviral symbols are as in FIG. 27A-F “NT”, not tested.
  • SIV Simian immunodeficiency virus
  • TM fusion protein DP178-like region antiviral data
  • FIG. 49A-L DP178-derived peptide antiviral data [(SEQ ID NO:158); T50:(SEQ ID NO:159); (SEQ ID NO:160); T234:(SEQ ID NO:161); T235:(SEQ ID NO:162); T570:(SEQ ID NO:163); T381:(SEQ ID NO:164); T677:(SEQ ID NO:165); T589:(SEQ ID NO:166); T590:(SEQ ID NO:167); T591:(SEQ ID NO:168); T270:(SEQ ID NO:169); T271:(SEQ ID NO:170); T273:(SEQ ID NO:171); T608:(SEQ ID NO:172); T609:(SEQ ID NO:173); T610:(SEQ ID NO:174); T611:(SEQ ID NO:175); T612:(SEQ ID NO:176); T595:(SEQ ID NO:177);
  • FIG. 49A-D The column to the immediate right of the name of the test peptide indicates the size of the test peptide and points out whether the peptide is derived from a one amino acid peptide “walk” across the DP178 region.
  • the next column to the right indicates whether the test peptide contains a point mutation, while the column to its right indicates whether certain amino acid residues have been added to or removed from the DP178-derived amino acid sequence.
  • FIG. 49E-H The column to the immediate right of the test peptide name indicates whether the peptide represents a DP178 truncation, the next column to the right points out whether the peptide contains a point mutation, and the column to its right indicates whether the peptide contains amino acids which have been added to or removed from the DP178 sequence itself FIG.
  • IC 50 is as defined in FIG. 27A-F , and IC 50 values were obtained using purified peptides except where marked with an asterisk (*), in which case the IC 50 was obtained using a crude peptide preparation.
  • FIG. 50A-B DP107 and DP107 gp41 region truncated peptide antiviral data (SEQ ID NO:201).
  • IC 50 as defined in FIG. 27A-D , and IC 50 values were obtained using purified peptides except where marked with an asterisk (*), in which case the IC 50 was obtained using a crude peptide preparation.
  • FIG. 51A-C Epstein-Barr virus Strain B95-8 BZLF1 DP178/DP107 analog region peptide walks and electrophoretic mobility shift assay results.
  • amino acid residue within this region which corresponds to the first amino acid residue of each peptide is listed to the left of each peptide, while the amino acid residue within this region which corresponds to the last amino acid residue of each peptide is listed to the right of each peptide.
  • the length of each test peptide is listed at the far right of each line, under the heading “Res”.
  • ACT refers to a test peptide's ability to inhibit Zebra binding to its response element.
  • + refers to a visible, but incomplete, abrogation of the response element/Zebra homodimer complex;
  • +++ refers to a complete abrogation of the complex; and
  • represents a lack of complex disruption.
  • FIG. 52A-B Hepatitis B virus subtype AYW major surface antigen precursor S protein DP178/DP107 analog region and peptide walks.
  • 52 A depicts Domain I (SEQ ID NO:208) (S protein amino acid residues 174-219), which contains a potential DP178/DP107 analog region.
  • peptides are listed which represent one amino acid peptide “walks” through domain I.
  • 52 B depicts Domain II (SEQ ID NO:209) (S protein amino acid residues 233-290), which contains a second potential DP178/DP107 analog region.
  • peptides are listed which represent one amino acid peptide “walks” through domain II.
  • peptides which may exhibit antifusogenic activity, antiviral capability, and/or the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • the peptides described include, first, DP178 (SEQ ID NO:1), a gp41-derived 36 amino acid peptide and fragments and analogs of DP178.
  • DP107 is a 38 amino acid peptide corresponding to residues 558 to 595 of the HIV-1 LAI transmembrane (TM) gp41 protein.
  • TM HIV-1 LAI transmembrane
  • Such DP107 analogs may exhibit antifusogenic capability, antiviral activity or an ability to modulate intracellular processes involving coiled-coil structures.
  • peptides of the invention include DP107 and DP178 are described herein having amino acid sequences recognized by the 107 ⁇ 178 ⁇ 4, ALLMOTI5, and PLZIP search motifs. Such motifs are also discussed.
  • peptides of the invention are antifusogenic, antiviral, intracellular modulatory, and diagnostic uses of the peptides of the invention. Further, procedures are described for the use of the peptides of the invention for the identification of compounds exhibiting antifusogenic, antiviral or intracellular modulatory activity.
  • DP178 corresponds to a putative ⁇ -helix region located in the C-terminal end of the gp41 ectodomain, and appears to associate with a distal site on gp41 whose interactive structure is influenced by the leucine zipper motif, a coiled-coil structure, referred to as DP107.
  • the association of these two domains may reflect a molecular linkage or “molecular clasp” intimately involved in the fusion process.
  • mutations in the C-terminal ⁇ -helix motif of gp41 tend to enhance the fusion ability of gp41, whereas mutations in the leucine zipper region (i.e., the DP107 domain) decrease or abolish the fusion ability of the viral protein. It may be that the leucine zipper motif is involved in membrane fusion while the C-terminal ⁇ -helix motif serves as a molecular safety to regulate the availability of the leucine zipper during virus-induced membrane fusion.
  • FIG. 11A-B Two models are proposed of gp41-mediated membrane fusion which are schematically shown in FIG. 11A-B .
  • the reason for proposing two models is that the temporal nature of the interaction between the regions defined by DP107 and DP178 cannot, as yet, be pinpointed.
  • Each model envisions two conformations for gp41-one in a “native” state as it might be found on a resting virion. The other in a “fusogenic” state to reflect conformational changes triggered following binding of gp120 to CD4 and just prior to fusion with the target cell membrane.
  • the strong binding affinity between gp120 and CD4 may actually represent the trigger for the fusion process obviating the need for a pH change such as occurs for viruses that fuse within intracellular vesicles.
  • the two major features of both models are: (1) the leucine zipper sequences (DP107) in each chain of oligomeric enveloped are held apart in the native state and are only allowed access to one another in the fusogenic state so as to form the extremely stable coiled-coils, and (2) association of the DP178 and DP107 sites as they exist in gp41 occur either in the native or fusogenic state.
  • FIG. 11A depicts DP178/DP107 interaction in the native state as a molecular clasp.
  • the model in FIG. 11B can be considered.
  • both DP107 and DP178 are potent inhibitors of HIV infection and fusion, probably by virtue of their ability to form complexes with viral gp41 and interfere with its fusogenic process; e.g., during the structural transition of the viral protein from the native structure to the fusogenic state, the DP178 and DP107 peptides may gain access to their respective binding sites on the viral gp41, and exert a disruptive influence.
  • DP107 peptides which demonstrate anti-HIV activity are described in Applicants' co-pending application Ser. No. 08/264,531, filed Jun. 23, 1994, which is incorporated by reference herein in its entirety.
  • a truncated recombinant gp41 protein corresponding to the ectodomain of gp41 containing both DP107 and DP178 domains did not inhibit HIV-1 induced fusion.
  • the inactive recombinant protein was transformed to an active inhibitor of HIV-1 induced fusion. This transformation may result from liberation of the potent DP178 domain from a molecular clasp with the leucine zipper, DP107 domain.
  • the DP178 peptide (SEQ ID NO:1) of the invention corresponds to amino acid residues 638 to 673 of the transmembrane protein gp41 from the HIV-1 LAI isolate, and has the 36 amino acid sequence (reading from amino to carboxy terminus):
  • the peptides of the invention may include truncations of the DP178 (SEQ ID NO:1) peptide which exhibit antifusogenic activity, antiviral activity and/or the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • Truncations of DP178 (SEQ ID NO:1) peptides may comprise peptides of between 3 and 36 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 36-mer polypeptide), as shown in Tables I and IA, below. Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus.
  • X may represent an amino group (—NH 2 ) and “Z” may represent a carboxyl (—COOH) group.
  • X may represent a hydrophobic group, including but not limited to carbobenzyl, dansyl, or T-butoxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group.
  • Z may represent an amido group; a T-butoxycarbonyl group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group.
  • a preferred “X” or “Z” macromolecular group is a peptide group.
  • “X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • X may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • Z may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • the peptides of the invention also include DP178-like peptides.
  • DP178-like refers, first, to DP178 and DP178 truncations which contain one or more amino acid substitutions, insertions and/or deletions.
  • DP-178-like refers to peptide sequences identified or recognized by the ALLMOTI5, 107 ⁇ 178 ⁇ 4 and PLZIP search motifs described herein, having structural and/or amino acid motif similarity to DP178.
  • the DP178-like peptides of the invention may exhibit antifusogenic or antiviral activity, or may exhibit the ability to modulate intracellular processes involving coiled-coil peptides. Further, such DP178-like peptides may possess additional advantageous features, such as, for example, increased bioavailability, and/or stability, or reduced host immune recognition.
  • HIV-1 and HIV-2 enveloped proteins are structurally distinct, but there exists a striking amino acid conservation within the DP178-corresponding regions of HIV-1 and HIV-2.
  • the amino acid conservation is of a periodic nature, suggesting some conservation of structure and/or function. Therefore, one possible class of amino acid substitutions would include those amino acid changes which are predicted to stabilize the structure of the DP178 peptides of the invention. Utilizing the DP178 and DP178 analog sequences described herein, the skilled artisan can readily compile DP178 consensus sequences and ascertain from these, conserved amino acid residues which would represent preferred amino acid substitutions.
  • the amino acid substitutions may be of a conserved or non-conserved nature.
  • conserved amino acid substitutions consist of replacing one or more amino acids of the DP178 (SEQ ID NO:1) peptide sequence with amino acids of similar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to aspartic acid (D) amino acid substitution.
  • Non-conserved substitutions consist of replacing one or more amino acids of the DP178 (SEQ ID NO:1) peptide sequence with amino acids possessing dissimilar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to valine (V) substitution.
  • Amino acid insertions may consist of single amino acid residues or stretches of residues.
  • the insertions may be made at the carboxy or amino terminal end of the DP178 or DP178 truncated peptides, as well as at a position internal to the peptide.
  • Such insertions will generally range from 2 to 15 amino acids in length. It is contemplated that insertions made at either the carboxy or amino terminus of the peptide of interest may be of a broader size range, with about 2 to about 50 amino acids being preferred.
  • One or more such insertions may be introduced into DP178 (SEQ.
  • Preferred amino or carboxy terminal insertions are peptides ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP178 gp41 amino acid sequence, respectively.
  • a preferred amino terminal or carboxy terminal amino acid insertion would contain gp41 amino acid sequences found immediately amino to or carboxy to the DP178 region of the gp41 protein.
  • DP178 SEQ ID NO:1
  • DP178 truncations are also within the scope of the invention. Such deletions consist of the removal of one or more amino acids from the DP178 or DP178-like peptide sequence, with the lower limit length of the resulting peptide sequence being 4 to 6 amino acids. Such deletions may involve a single contiguous or greater than one discrete portion of the peptide sequences.
  • deletions may be introduced into DP178 (SEQ ID NO:1) or DP178 truncations, as long as such deletions result in peptides which may still be recognized by the 107 ⁇ 178 ⁇ 4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • peptides of the invention include peptides having amino acid sequences corresponding to DP107 analogs.
  • DP107 is a 38 amino acid peptide which exhibits potent antiviral activity, and corresponds to residues 558 to 595 of HIV-1 LAI transmembrane (TM) gp41 protein, as shown here:
  • the peptides of the invention may include truncations of the DP107 peptide which exhibit antifusogenic activity, antiviral activity and/or the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • Truncations of DP107 peptides may comprise peptides of between 3 and 38 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 38-mer polypeptide), as shown in Tables II and IIA, below. Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus.
  • X may represent an amino group (—NH 2 ) and “Z” may represent a carboxyl (—COOH) group.
  • X may represent a hydrophobic group, including but not limited to carbobenzyl, dansyl, or T-butoxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group.
  • Z may represent an amido group; a T-butoxycarbonyl group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group.
  • a preferred “X” or “Z” macromolecular group is a peptide group.
  • X may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • Z may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • X may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • Z may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • the peptides of the invention also include DP107-like peptides.
  • DP107-like refers, first, to DP107 and DP107 truncations which contain one or more amino acid substitutions, insertions and/or deletions.
  • DP-107-like refers to peptide sequences identified or recognized by the ALLMOTI5, 107 ⁇ 178 ⁇ 4 and PLZIP search motifs described herein, having structural and/or amino acid motif similarity to DP107.
  • the DP107-like peptides of the invention may exhibit antifusogenic or antiviral activity, or may exhibit the ability to modulate intracellular processes involving coiled-coil peptides. Further, such DP107-like peptides may possess additional advantageous features, such as, for example, increased bioavailability, and/or stability, or reduced host immune recognition.
  • HIV-1 and HIV-2 enveloped proteins are structurally distinct, but there exists a striking amino acid conservation within the DP107-corresponding regions of HIV-1 and HIV-2.
  • the amino acid conservation is of a periodic nature, suggesting some conservation of structure and/or function. Therefore, one possible class of amino acid substitutions would include those amino acid changes which are predicted to stabilize the structure of the DP107 peptides of the invention. Utilizing the DP107 and DP107 analog sequences described herein, the skilled artisan can readily compile DP107 consensus sequences and ascertain from these, conserved amino acid residues which would represent preferred amino acid substitutions.
  • the amino acid substitutions may be of a conserved or non-conserved nature.
  • conserved amino acid substitutions consist of replacing one or more amino acids of the DP107 peptide sequence with amino acids of similar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to aspartic acid (D) amino acid substitution.
  • Non-conserved substitutions consist of replacing one or more amino acids of the DP107 peptide sequence with amino acids possessing dissimilar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to valine (V) substitution.
  • Amino acid insertions may consist of single amino acid residues or stretches of residues.
  • the insertions may be made at the carboxy or amino terminal end of the DP107 or DP107 truncated peptides, as well as at a position internal to the peptide.
  • Such insertions will generally range from 2 to 15 amino acids in length. It is contemplated that insertions made at either the carboxy or amino terminus of the peptide of interest may be of a broader size range, with about 2 to about 50 amino acids being preferred.
  • One or more such insertions may be introduced into DP107 or DP107 truncations, as long as such insertions result in peptides which may still be recognized by the 107 ⁇ 178 ⁇ 4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • Preferred amino or carboxy terminal insertions are peptides ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP107 gp41 amino acid sequence, respectively.
  • a preferred amino terminal or carboxy terminal amino acid insertion would contain gp41 amino acid sequences found immediately amino to or carboxy to the DP107 region of the gp41 protein.
  • Deletions of DP107 or DP178 truncations are also within the scope of the invention. Such deletions consist of the removal of one or more amino acids from the DP107 or DP107-like peptide sequence, with the lower limit length of the resulting peptide sequence being 4 to 6 amino acids. Such deletions may involve a single contiguous or greater than one discrete portion of the peptide sequences.
  • deletions may be introduced into DP107 or DP107 truncations, as long as such deletions result in peptides which may still be recognized by the 107 ⁇ 178 ⁇ 4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • Peptides corresponding to analogs of the DP178, DP178 truncations, DP107 and DP107 truncation sequences of the invention, described, above, in Sections 5.1 and 5.2 may be found in other viruses, including, for example, non-HIV-1 LAI enveloped viruses, non-enveloped viruses and other non-viral organisms.
  • analog refers to a peptide which is recognized or identified via the 107 ⁇ 178 ⁇ 4, ALLMOTI5 and/or PLZIP search strategies discussed below. Further, such peptides may exhibit antifusogenic capability, antiviral activity, or the ability to modulate intracellular processes involving coiled-coil structures.
  • Such DP178 and DP107 analogs may, for example, correspond to peptide sequences present in TM proteins of enveloped viruses and may, additionally correspond to peptide sequences present in non enveloped and non-viral organisms.
  • Such peptides may exhibit anti fusogenic activity, antiviral activity, most particularly antiviral activity which is specific to the virus in which their native sequences are found, or may exhibit an ability to modulate intracellular processes involving coiled-coil peptide structures.
  • DP178 analogs are peptides whose amino acid sequences are comprised of the amino acid sequences of peptide regions of, for example, other (i.e., other than HIV-1 LAI ) viruses that correspond to the gp41 peptide region from which DP178 (SEQ ID:1) was derived. Such viruses may include, but are not limited to, other HIV-1 isolates and HIV-2 isolates.
  • DP178 analogs derived from the corresponding gp41 peptide region of other (i.e., non HIV-1 LAI ) HIV-1 isolates may include, for example, peptide sequences as shown below.
  • SEQ ID NO:3 (DP-185; SEQ ID NO: 3) NH 2 -YT NT I YT L L EESQNQQEKNEQELLELDKWASLWNWF-COOH; (SEQ ID NO: 4) NH 2 -YT GI I YN L L EESQNQQEKNEQELLELDKWA N LWNWF-COOH; (SEQ ID NO: 5) NH 2 -YTSLI Y SL L E K SQIQQEKNEQELLELDKWASLWNWF-COOH.
  • SEQ ID NO:3 (DP-185), SEQ ID NO:4, and SEQ ID NO:5 are derived from HIV-1 SF2 , HIV-1 RF , and HIV-1 MN isolates, respectively.
  • Underlined amino acid residues refer to those residues that differ from the corresponding position in the DP178 (SEQ ID NO:1) peptide.
  • One such DP178 analog, DP-185 (SEQ ID NO:3) is described in the Example presented in Section 6, below, where it is demonstrated that DP-185 (SEQ ID NO:3) exhibits antiviral activity.
  • the DP178 analogs of the invention may also include truncations, as described above. Further, the analogs of the invention modifications such those described for DP178 analogs in Section 5.1., above.
  • the DP178 analogs of the invention represent peptides whose amino acid sequences correspond to the DP178 region of the gp41 protein, it is also contemplated that the peptides of the invention may, additionally, include amino sequences, ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP178 amino acid sequence.
  • a DP178 analog derived from the HIV-2 NIHZ isolate has the 36 amino acid sequence (reading from amino to carboxy terminus):
  • Table III and Table IV show some possible truncations of the HIV-2 NIHZ DP178 analog, which may comprise peptides of between 3 and 36 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 36-mer polypeptide). Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus. “X” may represent an amino group (—NH 2 ) and “Z” may represent a carboxyl (—COOH) group.
  • “X” may represent a hydrophobic group, including but not limited to carbobenzyl, dansyl, or T-butoxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group.
  • “Z” may represent an amido group; a T-butoxycarbonyl group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group.
  • a preferred “X” or “Z” macromolecular group is a peptide group.
  • X may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • Z may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • X may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • Z may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • DP178 and DP107 analogs are recognized or identified, for example, by utilizing one or more of the 107 ⁇ 178 ⁇ 4, ALLMOTI5 or PLZIP computer-assisted search strategies described and demonstrated, below, in the Examples presented in Sections 9 through 16 and 19 through 25.
  • the search strategy identifies additional peptide regions which are predicted to have structural and/or amino acid sequence features similar to those of DP107 and/or DP178.
  • search strategies are described fully, below, in the Example presented in Section 9. While this search strategy is based, in part, on a primary amino acid motif deduced from DP107 and DP178, it is not based solely on searching for primary amino acid sequence homologies, as such protein sequence homologies exist within, but not between major groups of viruses.
  • primary amino acid sequence homology is high within the TM protein of different strains of HIV-1 or within the TM protein of different isolates of simian immunodeficiency virus (SIV).
  • SIV simian immunodeficiency virus
  • Primary amino acid sequence homology between HIV-1 and SIV is low enough so as not to be useful. It is not possible, therefore, to find peptide regions similar to DP107 or DP178 within other viruses, or within non-viral organisms, whether structurally, or otherwise, based on primary sequence homology, alone.
  • Lupas search algorithm is based on the structure of coiled coils from proteins that are not structurally or functionally similar to the TM proteins of viruses, antiviral peptides (e.g. DP107 and DP178) of which are an object of this invention.
  • the computer search strategy of the invention as demonstrated in the Examples presented below, in Sections 9 through 16 and 19 through 25, successfully identifies regions of proteins similar to DP107 or DP178.
  • This search strategy was designed to be used with a commercially-available sequence database package, preferably PC/Gene.
  • search motifs the 107 ⁇ 178 ⁇ 4, ALLMOTI5 and PLZIP motifs, were designed and engineered to range in stringency from strict to broad, as discussed in this Section and in Section 9, with 107 ⁇ 178 ⁇ 4 being preferred.
  • the sequences identified via such search motifs such as those listed in Tables V-XIV, below, potentially exhibit antifusogenic, such as antiviral, activity, may additionally be useful in the identification of antifusogenic, such as antiviral, compounds, and are intended to be within the scope of the invention.
  • Coiled-coiled sequences are thought to consist of heptad amino acid repeats.
  • the amino acid positions within the heptad repeats are sometimes referred to as A through G, with the first position being A, the second B, etc.
  • the motifs used to identify DP107-like and DP178-like sequences herein are designed to specifically search for and identify such heptad repeats.
  • amino acids enclosed by brackets i.e., [ ] designate the only amino acid residues that are acceptable at the given position
  • amino acids enclosed by braces i.e., ⁇ ⁇ , designate the only amino acids which are unacceptable at the given heptad position.
  • bracketed or braced amino acids When a set of bracketed or braced amino acids is followed by a number in parentheses i.e., ( ), it refers to the number of subsequent amino acid positions for which the designated set of amino acids hold, e.g, a (2) means “for the next two heptad amino acid positions”.
  • ALLMOTI5 motif it may also be designed to search for 28-mers, by only repeating the initial motif four times. With respect to the ALLMOTI5 motif, a 35-mer search is preferred. Those viral (non-bacteriophage) sequences identified via such an ALLMOTI5 motif are listed in Table V, below, at the end of this Section. The viral sequences listed in Table V potentially exhibit antiviral activity, may be useful in the identification of antiviral compounds, and are intended to be within the scope of the invention. In those instances wherein a single gene exhibits greater than one sequence recognized by the ALLMOTI5 search motif, the amino a cid residue numbers of these sequences are listed under “Area 2”, Area 3”, etc. This convention is used for each of the Tables listed, below, at the end of this Section.
  • the 107 ⁇ 178 ⁇ 4 motif is written as follows:
  • This motif is designed to search for four consecutive heptad repeats (thus the repeat of the first line four times), meaning that it searches for 28-mer sized peptides. It may also be designed to search for 35-mers, by repeating the initial motif five times. With respect to the 107 ⁇ 178 ⁇ 4 motif, a 28-mer search is preferred.
  • the 107 ⁇ 178 ⁇ 4 search motif was also utilized to identify non-viral procaryotic protein sequences, as listed in Table VIII, below, at the end of this Section. Further, this search motif was used to reveal a number of human proteins. The results of this human protein 107 ⁇ 178 ⁇ 4 search is listed in Table IX, below, at the end of this Section. The sequences listed in Tables VIII and IX, therefore, reveal peptides which may be useful as antifusogenic compounds or in the identification of antifusogenic compounds, and are intended to be within the scope of the invention.
  • the PLZIP series of motifs are as listed in FIG. 19 . These motifs are designed to identify leucine zipper coiled-coil like heptads wherein at least one proline residue is present at some predefined distance N-terminal to the repeat. These PLZIP motifs find regions of proteins with similarities to HIV-1 DP178 generally located just N-terminal to the transmembrane anchor. These motifs may be translated according to the same convention described above. Each line depicted in FIG. 19 represents a single, complete search motif. “X” in these motifs refers to any amino acid residue. In instances wherein a motif contains two numbers within parentheses, this refers to a variable number of amino acid residues. For example, X (1,12) is translated to “the next one to twelve amino acid residues, inclusive, may be any amino acid”.
  • Tables X through XIV below, at the end of this Section, list sequences identified via searches conducted with such PLZIP motifs. Specifically, Table X lists viral sequences identified via PCTLZIP, P1CTLZIP and P2CTLZIP search motifs, Table XI lists viral sequences identified via P3CTLZIP, P4CTLZIP, P5CTLZIP and P6CTLZIP search motifs, Table XII lsts viral sequences identified via P7CTLZIP, P8CTLZIP and P9CTLZIP search motifs, Table XIII lists viral sequences identified via P12LZIPC searches and Table XIV lists viral sequences identified via P23TLZIPC search motifs The viral sequences listed in these tables represent peptides which potentially exhibit antiviral activity, may be useful in the identification of antiviral compounds, and are intended to be within the scope of the invention.
  • Example presented in Sections 17, 18, 26 and 27 below demonstrate that viral sequences identified via the motif searches described herein identify substantial antiviral characteristics. Specifically, the Example presented in Section 17 describes peptides with anti-respiratory syncytial virus activity, the Example presented in Section 18 describes peptides with anti-parainfluenza virus activity, the Example presented in Section 26 describes peptides with anti-measles virus activity and the Example presented in Section 27 describes peptides with anti-simian immunodeficiency virus activity.
  • the DP107 and DP178 analogs may, further, contain any of the additional groups described for DP178, above, in Section 5.1.
  • these peptides may include any of the additional amino-terminal groups as described above for “X” groups, and may also include any of the carboxy-terminal groups as described, above, for “Z” groups.
  • truncations of the identified DP107 and DP178 peptides are among the peptides of the invention. Further, such DP107 and DP178 analogs and DP107/DP178 analog truncations may exhibit one or more amino acid substitutions, insertion, and/or deletions.
  • the DP178 analog amino acid substitutions, insertions and deletions are as described, above, for DP178-like peptides in Section 5.1.
  • the DP-107 analog amino acid substitutions, insertions and deletions are also as described, above, for DP107-like peptides in Section 5.2.
  • Tables XV through XXII present representative examples of such DP107/DP178 truncations. Specifically, Table XV presents Respiratory Syncytial Virus F1 region DP107 analog carboxy truncations, Table XVI presents Respiratory Syncytial Virus F1 region DP107 analog amino truncations, Table XVII presents Respiratory Syncytial Virus F1 region DP178 analog carboxy truncations, Table XVIII presents Respiratory Syncytial Virus F1 region DP178 analog amino truncations, Table XIX presents Human Parainfluenza Virus 3 F1 region DP178 analog carboxy truncations, Table XX presents Human Parainfluenza Virus 3 F1 region DP178 analog amino truncations, Table XXI presents Human Parainfluenza Virus 3 F1 region DP107 analog carboxy truncations and Table XXII presents Human Parainfluenza Virus 3 F1 region
  • Table XXIII presents DP107/DP178 analogs and analog truncations which exhibit substantial antiviral activity. These antiviral peptides are grouped according to the specific virus which they inhibit, including respiratory syncytial virus, human parainfluenza virus 3, simian immunodeficiency virus and measles virus.
  • the peptides of the invention may be synthesized or prepared by techniques well known in the art. See, for example, Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman and Co., NY, which is incorporated herein by reference in its entirety. Short peptides, for example, can be synthesized on a solid support or in solution. Longer peptides may be made using recombinant DNA techniques.
  • the nucleotide sequences encoding the peptides of the invention may be synthesized, and/or cloned, and expressed according to techniques well known to those of ordinary skill in the art. See, for example, Sambrook, et al., 1989, Molecular Cloning, A Laboratory Manual, Vols. 1-3, Cold Spring Harbor Press, NY.
  • the peptides of the invention may alternatively be synthesized such that one or more of the bonds which link the amino acid residues of the peptides are non-peptide bonds. These alternative non-peptide bonds may be formed by utilizing reactions well known to those in the art, and may include, but are not limited to imino, ester, hydrazide, semicarbazide, and azo bonds, to name but a few.
  • peptides comprising the sequences described above may be synthesized with additional chemical groups present at their amino and/or carboxy termini, such that, for example, the stability, bioavailability, and/or inhibitory activity of the peptides is enhanced.
  • hydrophobic groups such as carbobenzoxyl, dansyl, or t-butyloxycarbonyl groups, may be added to the peptides' amino termini.
  • an acetyl group or a 9-fluorenylmethoxy-carbonyl group may be placed at the peptides' amino termini.
  • the hydrophobic group, t-butyloxycarbonyl, or an amido group may be added to the peptides' carboxy termini. (See “Z” in Tables I to IV, above.)
  • the peptides of the invention may be synthesized such that their steric configuration is altered.
  • the D-isomer of one or more of the amino acid residues of the peptide may be used, rather than the usual L-isomer.
  • amino acid residues of the peptides of the invention may be substituted by one of the well known non-naturally occurring amino acid residues. Alterations such as these may serve to increase the stability, bioavailability and/or inhibitory action of the peptides of the invention.
  • any of the peptides described above may, additionally, have a macromolecular carrier group covalently attached to their amino and/or carboxy termini.
  • Such macromolecular carrier groups may include, for example, lipid-fatty acid conjugates, polyethylene glycol, carbohydrates or additional peptides.
  • “X”, in Tables I to IV, above, may therefore additionally represent any of the above macromolecular carrier groups covalently attached to the amino terminus of a peptide, with an additional peptide group being preferred.
  • “Z”, in Tables I to IV may additionally represent any of the macromolecular carrier groups described above.
  • Assays for cell fusion events are well known to those of skill in the art, and may be used in conjunction, for example, with the peptides of the invention to test the peptides' antifusogenic capabilities.
  • Cell fusion assays are generally performed in vitro. Such an assay may comprise culturing cells which, in the absence of any treatment would undergo an observable level of syncytial formation. For example, uninfected cells may be incubated in the presence of cells chronically infected with a virus that induces cell fusion.
  • viruses may include, but are not limited to, HIV, SIV, or respiratory syncytial virus.
  • cells are incubated in the presence of a peptide to be assayed.
  • a range of peptide concentrations may be tested. This range should include a control culture wherein no peptide has been added.
  • Standard conditions for culturing cells are used. After incubation for an appropriate period (24 hours at 37° C., for example) the culture is examined microscopically for the presence of multinucleated giant cells, which are indicative of cell fusion and syncytial formation.
  • Well known stains such as crystal violet stain, may be used to facilitate the visualization of syncytial formation.
  • the antiviral activity exhibited by the peptides of the invention may be measured, for example, by easily performed in vitro assays, such as those described below, which can test the peptides' ability to inhibit syncytia formation, or their ability to inhibit infection by cell-free virus. Using these assays, such parameters as the relative antiviral activity of the peptides, exhibit against a given strain of virus and/or the strain specific inhibitory activity of the peptide can be determined.
  • a cell fusion assay may be utilized to test the peptides' ability to inhibit viral-induced, such as HIV-induced, syncytia formation in vitro.
  • Such an assay may comprise culturing uninfected cells in the presence of cells chronically infected with a syncytial-inducing virus and a peptide to be assayed. For each peptide, a range of peptide concentrations may be tested. This range should include a control culture wherein no peptide has been added. Standard conditions for culturing, well known to those of ordinary skill in the art, are used.
  • the culture After incubation for an appropriate period (24 hours at 37° C., for example) the culture is examined microscopically for the presence of multinucleated giant cells, which are indicative of cell fusion and syncytia formation.
  • Well known stains such as crystal violet stain, may be used to facilitate syncytial visualization.
  • an assay would comprise CD-4 + cells (such as Molt or CEM cells, for example) cultured in the presence of chronically HIV-infected cells and a peptide to be assayed.
  • a reverse transcriptase (RT) assay may be utilized to test the peptides' ability to inhibit infection of CD-4 + cells by cell-free HIV.
  • a reverse transcriptase (RT) assay may comprise culturing an appropriate concentration (i.e., TCID 50 ) of virus and CD-4 + cells in the presence of the peptide to be tested. Culture conditions well known to those in the art are used. As above, a range of peptide concentrations may be used, in addition to a control culture wherein no peptide has been added.
  • a cell-free supernatant is prepared, using standard procedures, and tested for the present of RT activity as a measure of successful infection.
  • the RT activity may be tested using standard techniques such as those described by, for example, Goff et al. (Goff, S. et al., 1981, J. Virol. 38:239-248) and/or Willey et al. (Willey, R. et al., 1988, J. Virol. 62:139-147). These references are incorporated herein by reference in their entirety.
  • In vivo assays may also be utilized to test, for example, the antiviral activity of the peptides of the invention.
  • To test for anti-HIV activity for example, the in vivo model described in Barnett et al. (Barnett, S. W. et al., 1994, Science 266:642-646) may be used.
  • anti-RSV activity can be assayed in vivo via well known mouse models.
  • RSV can be administered intranasally to mice of various inbred strains. Virus replicates in lungs of all strains, but the highest titers are obtained in P/N, C57L/N and DBA/2N mice. Infection of BALB/c mice produces an asymptomatic bronchiolitis characterized by lymphocytic infiltrates and pulmonary virus titers of 10 4 to 10 5 pfu/g of lung tissue (Taylor, G. et al., 1984, Infect. Immun. 43:649-655).
  • Cotton rat models of RSV are also well known. Virus replicates to high titer in the nose and lungs of the cotton rat but produces few if any signs of inflammation.
  • the peptides of the invention may be utilized as antifusogenic or antiviral compounds, or as compounds which modulate intracellular processes involving coiled coil peptide structures. Further, such peptides may be used to identify agents which exhibit antifusogenic, antiviral or intracellular modulatory activity. Still further, the peptides of the invention may be utilized as organism or viral type/subtype-specific diagnostic tools.
  • the antifusogenic capability of the peptides of the invention may additionally be utilized to inhibit or treat/ameliorate symptoms caused by processes involving membrane fusion events. Such events may include, for example, virus transmission via cell-cell fusion, abnormal neurotransmitter exchange via cell-fusion, and sperm-egg fusion. Further, the peptides of the invention may be used to inhibit free viral, such as retroviral, particularly HIV, transmission to uninfected cells wherein such viral infection involves membrane fusion events or involves fusion of a viral structure with a cell membrane.
  • free viral such as retroviral, particularly HIV
  • the intracellular disorders involving coiled coil peptides structures which may be ameliorated by the peptides of the invention are disorders involving, for example, bacterial toxins.
  • viruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to all strains of the viruses listed above, in Tables V through VII, and IX through XIV.
  • viruses include, for example, human retroviruses, particularly HIV-1 and HIV-2 and the human T-lymphocyte viruses (HTLV-I and II).
  • the non-human retroviruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to bovine leukosis virus, feline sarcoma and leukemia viruses, simian immunodeficiency, sarcoma and leukemia viruses, and sheep progress pneumonia viruses.
  • Non retroviral viruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to human respiratory syncytial virus, canine distemper virus, newcastle disease virus, human parainfluenza virus, influenza viruses, measles viruses, Epstein-Barr viruses, hepatitis B viruses, and simian Mason-Pfizer viruses.
  • Non enveloped viruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to picornaviruses such as polio viruses, hepatitis A virus, enterovirus, echoviruses and coxsackie viruses, papovaviruses such as papilloma virus, parvoviruses, adenoviruses and reoviruses.
  • picornaviruses such as polio viruses, hepatitis A virus, enterovirus, echoviruses and coxsackie viruses
  • papovaviruses such as papilloma virus, parvoviruses, adenoviruses and reoviruses.
  • DP107, DP178, DP107 analog and DP178 analog peptides form non-covalent protein-protein interactions which are required for normal activity of the virus.
  • the peptides of the invention may also be utilized as components in assays for the identification of compounds that interfere with such protein-protein interactions and may, therefore, act as antiviral agents. These assays are discussed, below, in Section 5.5.1.
  • the antiviral activity of the peptides of the invention may show a pronounced type and subtype specificity, i.e., specific peptides may be effective in inhibiting the activity of only specific viruses.
  • This feature of the invention presents many advantages.
  • One such advantage lies in the field of diagnostics, wherein one can use the antiviral specificity of the peptide of the invention to ascertain the identity of a viral isolate. With respect to HIV, one may easily determine whether a viral isolate consists of an HIV-1 or HIV-2 strain.
  • uninfected CD-4 + cells may be co-infected with an isolate which has been identified as containing HIV the DP178 (SEQ ID NO:1) peptide, after which the retroviral activity of cell supernatants may be assayed, using, for example, the techniques described above in Section 5.2.
  • Those isolates whose retroviral activity is completely or nearly completely inhibited contain HIV-1.
  • Those isolates whose viral activity is unchanged or only reduced by a small amount may be considered to not contain HIV-1.
  • Such an isolate may then be treated with one or more of the other DP178 peptides of the invention, and subsequently be tested for its viral activity in order to determine the identify of the viral isolate.
  • the DP107 and DP178 analogs of the invention may also be utilized in a diagnostic capacity specific to the type and subtype of virus or organism in which the specific peptide sequence is found.
  • a diagnostic procedure as described, above, for DP178, may be used in conjunction with the DP107/DP178 analog of interest.
  • DP107 and DP178 portions of the TM protein gp41 form non-covalent protein-protein interactions.
  • the maintenance of such interactions is necessary for normal viral infectivity.
  • compounds which bind DP107, bind DP178, and/or act to disrupt normal DP107/DP178 protein-protein interactions may act as antifusogenic, antiviral or cellular modulatory agents. Described below are assays for the identification of such compounds.
  • DP107 and DP178 peptides will be used as components of the assays described, but it is to be understood that any of the DP107 analog or DP178 analog peptides described, above, in Sections 5.1 through 5.3 may also be utilized as part of these screens for compounds.
  • Compounds which may be tested for an ability to bind DP107, DP178, and/or disrupt DP107/DP178 interactions, and which therefore, potentially represent antifusogenic, antiviral or intracellular modulatory compounds include, but are not limited to, peptides made of D- and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam, K. S. et al., 1991, Nature 354:82-84), phosphopeptides (in, for example, the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang, Z. et al., 1993, Cell 72:767-778), antibodies, and small organic or inorganic molecules. Synthetic compounds, natural products, and other sources of potentially effective materials may be screened in a variety of ways, as described in this Section.
  • the compounds, antibodies, or other molecules identified may be tested, for example, for an ability to inhibit cell fusion or viral activity, utilizing, for example, assays such as those described, above, in Section 5.5.
  • peptides which may be tested are soluble peptides comprising DP107 and/or DP178 domains, and peptides comprising DP107 and/or DP178 domains having one or more mutations within one or both of the domains, such as the M41-P peptide described, below, in the Example presented in Section 8, which contains a isoleucine to proline mutation within the DP178 sequence.
  • DP107-binding or DP178-binding compounds are assays which would include the attachment of either the DP107 or the DP178 peptide to a solid matrix, such as, for example, agarose or plastic beads, microtiter plate wells, petri dishes, or membranes composed of for example, nylon or nitrocellulose.
  • a solid matrix such as, for example, agarose or plastic beads, microtiter plate wells, petri dishes, or membranes composed of for example, nylon or nitrocellulose.
  • a solid matrix such as, for example, agarose or plastic beads, microtiter plate wells, petri dishes, or membranes composed of for example, nylon or nitrocellulose.
  • a solid matrix such as, for example, agarose or plastic beads, microtiter plate wells, petri dishes, or membranes composed of for example, nylon or nitrocellulose.
  • either the DP107 or DP178 protein may be anchored onto a solid surface, and the compound, or test substance, which is not anchored, is
  • an immobilized antibody preferably a monoclonal antibody, specific for the protein may be used to anchor the protein to the solid surface.
  • the surfaces may be prepared in advance and stored.
  • the labeled compound is added to the coated surface containing the anchored DP107 or DP178 peptide. After the reaction is complete, unreacted components are removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid surface.
  • the detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the compound is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed.
  • an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the compound (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).
  • such an assay can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for DP107 or DP178, whichever is appropriate for the given assay, or ab antibody specific for the compound, i.e., the test substance, in order to anchor any complexes formed in solution, and a labeled antibody specific for the other member of the complex to detect anchored complexes.
  • compounds may be screened for an ability to inhibit the formation of or, alternatively, disrupt DP107/DP178 complexes. Such compounds may then be tested for antifusogenic, antiviral or intercellular modulatory capability.
  • DP107 and DP178 will be referred to as “binding partners.” Compounds that disrupt such interactions may exhibit antiviral activity.
  • Such compounds may include, but are not limited to molecules such as antibodies, peptides, and the like described above.
  • the basic principle of the assay systems used to identify compounds that interfere with the interaction between the DP107 and DP178 peptides involves preparing a reaction mixture containing peptides under conditions and for a time sufficient to allow the two peptides to interact and bind, thus forming a complex.
  • the reaction is conducted in the presence and absence of the test compound, i.e., the test compound may be initially included in the reaction mixture, or added at a time subsequent to the addition of one of the binding partners; controls are incubated without the test compound or with a placebo.
  • the formation of any complexes between the binding partners is then detected.
  • the formation of a complex in the control reaction, but not in the reaction mixture containing the test compound indicates that the compound interferes with the interaction of the DP107 and DP178 peptides.
  • the assay for compounds that interfere with the interaction of the binding partners can be conducted in a heterogeneous or homogeneous format.
  • Heterogeneous assays involve anchoring one of the binding partners onto a solid phase and detecting complexes anchored on the solid phase at the end of the reaction.
  • homogeneous assays the entire reaction is carried out in a liquid phase.
  • the order of addition of reactants can be varied to obtain different information about the compounds being tested.
  • test compounds that interfere with the interaction between the binding partners e.g., by competition, can be identified by conducting the reaction in the presence of the test substance; i.e., by adding the test substance to the reaction mixture prior to or simultaneously with the binding partners.
  • test compounds that disrupt preformed complexes e.g. compounds with higher binding constants that displace one of the binding partners from the complex
  • test compounds that disrupt preformed complexes e.g. compounds with higher binding constants that displace one of the binding partners from the complex
  • one binding partner e.g., either the DP107 or DP178 peptide
  • a solid surface and its binding partner, which is not anchored, is labeled, either directly or indirectly.
  • the anchored species may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the protein and drying. Alternatively, an immobilized antibody specific for the protein may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.
  • the binding partner of the immobilized species is added to the coated surface with or without the test compound. After the reaction is complete, unreacted components are removed (e.g., by washing) and any complexes formed will remain immobilized on the solid surface.
  • the detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the binding partner was pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed.
  • an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the binding partner (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).
  • the antibody in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody.
  • test compounds which inhibit complex formation or which disrupt preformed complexes can be detected.
  • the reaction can be conducted in a liquid phase in the presence or absence of the test compound, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for one binding partner to anchor any complexes formed in solution, and a labeled antibody specific for the other binding partner to detect anchored complexes.
  • test compounds which inhibit complex or which disrupt preformed complexes can be identified.
  • a homogeneous assay can be used.
  • a preformed complex of the DP107 and DP178 peptides is prepared in which one of the binding partners is labeled, but the signal generated by the label is quenched due to complex formation (see, e.g., U.S. Pat. No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays).
  • the addition of a test substance that competes with and displaces one of the binding partners from the preformed complex will result in the generation of a signal above background. In this way, test substances which disrupt DP-107/DP-178 protein-protein interaction can be identified.
  • test compounds may be assayed for the their ability to disrupt a DP178/DP107 interaction, as measured immunometrically using an antibody specifically reactive to a DP107/DP178 complex (i.e., an antibody that recognizes neither DP107 nor DP178 individually).
  • an assay acts as a competition assay, and is based on techniques well known to those of skill in the art.
  • the above competition assay may be described, by way of example, and not by way of limitation, by using the DP178 and M41 ⁇ 178 peptides and by assaying test compounds for the disruption of the complexes formed by these two peptides by immunometrically visualizing DP178/M41 ⁇ 178 complexes via the human recombinant Fab, Fab-d, as described, below, in the Example presented in Section 8.
  • M41 ⁇ 178 is a maltose binding fusion protein containing a gp41 region having its DP178 domain deleted, and is described, below, in the Example presented in Section 8.
  • M41 ⁇ 178 may be immobilized onto solid supports such as microtiter wells. A series of dilutions of a test compound may then be added to each M41 ⁇ 178-containing well in the presence of a constant concentration of DP-178 peptide. After incubation, at, for example, room temperature for one hour, unbound DP-178 and test compound are removed from the wells and wells are then incubated with the DP178/M41 ⁇ 178-specific Fab-d antibody. After incubation and washing, unbound Fab-d is removed from the plates and bound Fab-d is quantitated. A no-inhibitor control should also be conducted. Test compounds showing an ability to disrupt DP178/M41 ⁇ 178 complex formation are identified by their concentration-dependent decrease in the level of Fab-d binding.
  • a variation of such an assay may be utilized to perform a rapid, high-throughput binding assay which is capable of directly measuring DP178 binding to M41 ⁇ 178 for the determination of binding constants of the ligand of inhibitory constants for competitors of DP178 binding.
  • Such an assay takes advantage of accepted radioligand and receptor binding principles. (See, for example, Yamamura, H. I. et al., 1985, “Neurotransmitter Receptor Binding”, 2nd ed., Raven Press, NY.)
  • M41 ⁇ 178 is immobilized onto a solid support such as a microtiter well.
  • DP178 binding to M41 ⁇ 178 is then quantitated by measuring the fraction of DP178 that is bound as 125 I-DP178 and calculating the total amount bound using a value for specific activity (dpm/ ⁇ g peptide) determined for each labeled DP178 preparation.
  • Specific binding to M41 ⁇ 178 is defined as the difference of the binding of the labeled DP178 preparation in the microtiter wells (totals) and the binding in identical wells containing, in addition, excess unlabeled DP178 (nonspecifics).
  • peptides of the invention may be administered using techniques well known to those in the art.
  • agents are formulated and administered systemically.
  • Techniques for formulation and administration may be found in “Remington's Pharmaceutical Sciences”, 18th ed., 1990, Mack Publishing Co., Easton, Pa.
  • Suitable routes may include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as, intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections, just to name a few.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • peptides of the invention or other inhibitory agents may be encapsulated into liposomes, then administered as described above.
  • Liposomes are spherical lipid bilayers with aqueous interiors. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior. The liposomal contents are both protected from the external microenvironment and, because liposomes fuse with cell membranes, are effectively delivered into the cell cytoplasm. Additionally, due to their hydrophobicity, when small molecules are to be administered, direct intracellular administration may be achieved.
  • Nucleotide sequences encoding the peptides of the invention which are to be intracellularly administered may be expressed in cells of interest, using techniques well known to those of skill in the art.
  • expression vectors derived from viruses such as retroviruses, vaccinia viruses, adeno-associated viruses, herpes viruses, or bovine papilloma viruses, may be used for delivery and expression of such nucleotide sequences into the targeted cell population. Methods for the construction of such vectors and expression constructs are well known.
  • peptides of the invention may be used as therapeutics in the treatment of AIDS.
  • the peptides may be used as prophylactic measures in previously uninfected individuals after acute exposure to an HIV virus.
  • prophylactic use of the peptides may include, but are not limited to, prevention of virus transmission from mother to infant and other settings where the likelihood of HIV transmission exists, such as, for example, accidents in health care settings wherein workers are exposed to HIV-containing blood products. The successful use of such treatments do not rely upon the generation of a host immune response directed against such peptides.
  • Effective dosages of the peptides of the invention to be administered may be determined through procedures well known to those in the art which address such parameters as biological half-life, bioavailability, and toxicity. Given the data presented below in Section 6, DP178, for example, may prove efficacious in vivo at doses required to achieve circulating levels of about 1 to about 10 ng per ml of peptide.
  • a therapeutically effective dose refers to that amount of the compound sufficient to result in amelioration of symptoms or a prolongation of survival in a patient.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Compounds which exhibit large therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (e.g., the concentration of the test compound which achieves a half-maximal inhibition of the fusogenic event, such as a half-maximal inhibition of viral infection relative to the amount of the event in the absence of the test compound) as determined in cell culture.
  • IC 50 e.g., the concentration of the test compound which achieves a half-maximal inhibition of the fusogenic event, such as a half-maximal inhibition of viral infection relative to the amount of the event in the absence of the test compound
  • levels in plasma may be measured, for example, by high performance liquid chromatography (HPLC).
  • the peptides of the invention may, further, serve the role of a prophylactic vaccine, wherein the host raises antibodies against the peptides of the invention, which then serve to neutralize HIV viruses by, for example, inhibiting further HIV infection.
  • Administration of the peptides of the invention as a prophylactic vaccine would comprise administering to a host a concentration of peptides effective in raising an immune response which is sufficient to neutralize HIV, by, for example, inhibiting HIV ability to infect cells.
  • concentration will depend upon the specific peptide to be administered, but may be determined by using standard techniques for assaying the development of an immune response which are well known to those of ordinary skill in the art.
  • the peptides to be used as vaccines are usually administered intramuscularly.
  • the peptides may be formulated with a suitable adjuvant in order to enhance the immunological response.
  • suitable adjuvants may include, but are not limited to mineral gels such as aluminum hydroxide; surface active substances such as lysolecithin, pluronic polyols, polyanions; other peptides; oil emulsions; and potentially useful human adjuvants such as BCG and Corynebacterium parvum .
  • Many methods may be used to introduce the vaccine formulations described here. These methods include but are not limited to oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, and intranasal routes.
  • an effective concentration of polyclonal or monoclonal antibodies raised against the peptides of the invention may be administered to a host so that no uninfected cells become infected by HIV.
  • concentration of such antibodies will vary according to each specific antibody preparation, but may be determined using standard techniques well known to those of ordinary skill in the art.
  • Administration of the antibodies may be accomplished using a variety of techniques, including, but not limited to those described in this section.
  • the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity, or to organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity).
  • the magnitude of an administrated dose in the management of the oncogenic disorder of interest will vary with the severity of the condition to be treated and the route of administration.
  • the dose and perhaps dose frequency will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
  • compositions of the present invention in particular, those formulated as solutions, may be administered parenterally, such as by intravenous injection.
  • the compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • the preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • compositions for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • DP178 (SEQ ID NO:1) is a Potent Inhibitor of HIV-1 Infection
  • DP178 (SEQ ID NO:1) is shown to be a potent inhibitor of HIV-1 mediated CD-4 + cell-cell fusion and infection by cell free virus.
  • this peptide completely blocks virus induced syncytia formation at concentrations of from 1-10 ng/ml.
  • the inhibitory concentration is somewhat higher, blocking infection at 90 ng/ml.
  • DP178 (SEQ ID NO:1) shows that the antiviral activity of DP178 (SEQ ID NO:1) is highly specific for HIV-1.
  • a synthetic peptide, DP-185 (SEQ ID NO:3), representing a HIV-1-derived DP178 homolog is also found to block HIV-1-mediated syncytia formation.
  • Peptides were synthesized using Fast Moc chemistry on an Applied Biosystems Model 431A peptide synthesizer. Generally, unless otherwise noted, the peptides contained amidated carboxy termini and acetylated amino termini. Amidated peptides were prepared using Rink resin (Advanced Chemtech) while peptides containing free carboxy termini were synthesized on Wang (p-alkoxy-benzyl-alcohol) resin (Bachem). First residues were double coupled to the appropriate resin and subsequent residues were single coupled. Each coupling step was followed by acetic anhydride capping.
  • Peptides were cleaved from the resin by treatment with trifluoracetic acid (TFA) (10 ml), H 2 O (0.5 ml), thioanisole (0.5 ml), ethanedithiol (0.25 ml), and crystalline phenol (0.75 g). Purification was carried out by reverse phase HPLC. Approximately 50 mg samples of crude peptide were chromatographed on a Waters Delta Pak C18 column (19 mm ⁇ 30 cm, 15 ⁇ spherical) with a linear gradient; H 2 O/acetonitrile 0.1% TFA. Lyophilized peptides were stored desiccated and peptide solutions were made in water at about 1 mg/ml.
  • TFA trifluoracetic acid
  • Electrospray mass spectrometry yielded the following results: DP178 (SEQ ID NO:1):4491.87 (calculated 4491.94); DP-180 (SEQ ID NO:2):4491.45 (calculated 4491.94); DP-185 (SEQ ID NO:3):not done (calculated 4546.97).
  • the HIV-1 LAI virus was obtained from R. Gallo (Popovic, M. et al., 1984, Science 224:497-508) and propagated in CEM cells cultured in RPMI 1640 containing 10% fetal calf serum. Supernatant from the infected CEM cells was passed through a 0.2 ⁇ m filter and the infectious titer estimated in a microinfectivity assay using the AA5 cell line to support virus replication. For this purpose, 250 of serial diluted virus was added to 75 ⁇ l AA5 cells at a concentration of 2 ⁇ 10 5 /ml in a 96-well microtitre plate. Each virus dilution was tested in triplicate. Cells were cultured for eight days by addition of fresh medium every other day.
  • TCID 50 was calculated according to the Reed and Muench formula (Reed, L. J. et al., 1938, Am. J. Hyg. 27:493-497).
  • Synthetic peptides were incubated at 37° C. with either 247 TCID 50 (for experiment depicted in FIG. 2 ), or 62 TCID 50 (for experiment depicted in FIG. 3 ) units of HIV-1 LAI virus or 25 TCID 50 units of HIV-2 NIHZ and CEM CD4 + cells at peptide concentrations of 0, 0.04, 0.4, 4.0, and 40 ⁇ g/ml for 7 days.
  • the resulting reverse transcriptase (RT) activity in counts per minute was determined using the assay described, below, in Section 6.1.5. See, Reed, L. J. et al., 1938, Am. J. Hyg. 27: 493-497 for an explanation of TCID 50 calculations.
  • RT micro-reverse transcriptase assay
  • Goff Goff, S. et al., 1981, J. Virol. 38:239-248
  • Willey et al. Willey, R. et al., 1988, J. Virol. 62:139-147.
  • Supernatants from virus/cell cultures are adjusted to 1% Triton-X100.
  • a 10 ⁇ l sample of supernatant was added to 50 ⁇ l of RT cocktail in a 96-well U-bottom microtitre plate and the samples incubated at 37° C. for 90 min.
  • the RT cocktail contained 75 mM KCl, 2 mM dithiothreitol, 5 mM MgCl 2 , 5 ⁇ g/ml poly A (Pharmacia, cat. No. 27-4110-01), 0.25 units/ml oligo dT (Pharmacia, cat. No. 27-7858-01), 0.05% NP40, 50 mM Tris-HCl, pH 7.8, 0.5 ⁇ M non-radioactive dTTP, and 10 ⁇ Ci/ml 32 P-dTTP (Amersham, cat. No. PB. 10167).
  • reaction mixture 40 ⁇ l was applied to a Schleicher and Schuell (S+S) NA45 membrane (or DE81 paper) saturated in 2 ⁇ SSC buffer (0.3M NaCl and 0.003M sodium citrate) held in a S+S Minifold over one sheet of GB003 (S+S) filter paper, with partial vacuum applied.
  • S+S Schleicher and Schuell
  • 2 ⁇ SSC buffer 0.M NaCl and 0.003M sodium citrate
  • DP178 SEQ ID NO:1 afforded complete protection against each of the HIV-1 isolates down to the lowest concentration of DP178 (SEQ ID NO:1) used.
  • the lowest concentration tested was 12.5 ng/ml; for all other HIV-1 viruses, the lowest concentration of DP178 (SEQ ID NO:1) used in this study was 100 ng/ml.
  • a second peptide, DP-180 (SEQ ID NO:2), containing the same amino acid residues as DP178 (SEQ ID NO:1) but arranged in a random order exhibited no evidence of anti-fusogenic activity even at the high concentration of 40 ⁇ g/ml ( FIG. 4 ).
  • DP178 SEQ ID NO:1 homolog for its ability to inhibit HIV-1-induced syncytia formation.
  • SEQ ID NO:3 the sequence of DP-185 (SEQ ID NO:3) is slightly different from DP178 (SEQ ID NO:1) in that its primary sequence is taken from the HIV-1 SF2 isolate and contains several amino acid differences relative to DP178 (SEQ ID NO:1) near the N terminus.
  • DP-185 SEQ ID NO:3
  • DP178 SEQ ID NO:1 HIV-1 and HIV-2 inhibitory activity.
  • DP178 SEQ ID NO:1 blocked HIV-1-mediated syncytia formation at peptide concentrations below 1 ng/ml.
  • DP178 SEQ ID NO:1 failed, however, to block HIV-2 mediated syncytia formation at concentrations as high as 10 ⁇ g/ml.
  • This striking 4 log selectivity of DP178 (SEQ ID NO:1) as an inhibitor of HIV-1-mediated cell fusion demonstrates an unexpected HIV-1 specificity in the action of DP178 (SEQ ID NO:1).
  • DP178 (SEQ ID NO:1) inhibition of HIV-1-mediated cell fusion, but the peptide's inability to inhibit HIV-2 medicated cell fusion in the same cell type at the concentrations tested provides further evidence for the high degree of selectivity associated with the antiviral action of DP178 (SEQ ID NO:1).
  • DP178 (SEQ ID:1) was next tested for its ability to block CD-4 + CEM cell infection by cell free HIV-1 virus.
  • the results, shown in FIG. 2 are from an experiment in which DP178 (SEQ ID NO:1) was assayed for its ability to block infection of CEM cells by an HIV-1 LAI isolate. Included in the experiment were three control peptides, DP-116 (SEQ ID NO:9), DP-125 (SEQ ID NO:8), and DP-118 (SEQ ID NO:10).
  • DP-116 (SEQ ID NO:9) represents a peptide previously shown to be inactive using this assay, and DP-125 (SEQ ID:8; Wild, C. et al., 1992, Proc. Natl. Acad.
  • DP-118 SEQ ID NO:10
  • DP178 SEQ ID NO:1
  • DP-125 SEQ: ID NO:8
  • DP-118 SEQ ID NO:10
  • DP178 SEQ ID NO:1
  • CEM cells CEM cells
  • 62 TCID 50 HIV-1 LAI or 25 GCID 50 HIV-2 NIHZ were used in these experiments, and were incubated for 7 days.
  • DP178 (SEQ ID NO:1) inhibited HIV-1 infection with an IC50 of about 31 ng/ml.
  • DP178 (SEQ ID NO:1) exhibited a much higher IC50 for HIV-2 NIHZ , thus making DP178 (SEQ ID NO:1) two logs more potent as a HIV-1 inhibitor than a HIV-2 inhibitor.
  • This finding is consistent with the results of the fusion inhibition assays described, above, in Section 6.2.1, and further supports a significant level of selectivity (i.e., for HIV-1 over HIV-2).
  • the 36 amino acid synthetic peptide inhibitor DP178 (SEQ ID NO:1) is shown to be non-cytotoxic to cells in culture, even at the highest peptide concentrations (40 ⁇ g/ml) tested.
  • Cell proliferation and toxicity assay Approximately 3.8 ⁇ 10 5 CEM cells for each peptide concentration were incubated for 3 days at 37° C. in T25 flasks. Peptides tested were DP178 (SEQ ID NO:1) and DP-116 (SEQ ID NO:9), as described in FIG. 1 . Peptides were synthesized as described, above, in Section 6.1. The concentrations of each peptide used were 0, 2.5, 10, and 40 ⁇ g/ml. Cell counts were taken at incubation times of 0, 24, 48, and 72 hours.
  • DP178 SEQ ID NO:1
  • DP-116 SEQ ID NO:9
  • DP178 SEQ ID NO:1 completely inhibits HIV-1 mediated syncytia formation at peptide concentrations between 1 and 10 ng/ml, and completely inhibits cell-free viral infection at concentrations of at least 90 ng/ml.
  • this study demonstrates that even at peptide concentrations greater than 3 log higher than the HIV inhibitory dose, DP178 (SEQ ID NO:1) exhibits no cytotoxic effects.
  • Soluble recombinant forms of gp41 used in the example described below provide evidence that the DP178 peptide associates with a distal site on gp41 whose interactive structure is influenced by the DP107 leucine zipper motif.
  • a single mutation disrupting the coiled-coil structure of the leucine zipper domain transformed the soluble recombinant gp41 protein from an inactive to an active inhibitor of HIV-1 fusion. This transformation may result from liberation of the potent DP178 domain from a molecular clasp with the leucine zipper, DP107, determinant.
  • the results also indicate that the anti-HIV activity of various gp41 derivatives (peptides and recombinant proteins) may be due to their ability to form complexes with viral gp41 and interfere with its fusogenic process.
  • gp41 sequence was amplified from pgtat (Malim et al., 1988, Nature 355: 181-183) by using polymerase chain reaction (PCR) with upstream primer 5′-ATGACGCTGACGGTACAGGCC-3′(SEQ ID NO:11) (primer A) and downstream primer 5′-TGACTAAGCTTAATACCACAGCCAATTTGTTAT-3′(SEQ ID NO:12) (primer B).
  • M41-P was constructed by using the T7-Gen in vitro mutagenesis kit from United States Biochemicals (USB) following the supplier's instructions.
  • the mutagenic primer (5′-GGAGCTGCTTGGGGCCCCAGAC-3′) (SEQ ID NO:13) introduces an Ile to Pro mutation in M41 at position 578.
  • M41 ⁇ 178 from which the DP-178 region has been deleted, was made by cloning the DNA fragment corresponding to gp41 amino acids 540-642 into the Xmn I site of pMal-p2.
  • Primer A and 5′-ATAGCTTCTAGATTAATTGTTAATTTCTCTGTCCC-3′(SEQ ID NO:15) (primer D) were used in the PCR with the template pgtat to generate the inserted DNA fragments.
  • M41-P was used as the template with primer A and D in PCR to generate M41-P ⁇ 178. All inserted sequences and mutated residues were checked by restriction enzyme analysis and confirmed by DNA sequencing.
  • the fusion proteins were purified according to the protocol described in the manufacturer's brochure of protein fusion and purification systems from New England Biolabs (NEB). Fusion proteins (10 ng) were analyzed by electrophoresis on 8% SDS polyacrylamide gels. Western blotting analysis was performed as described by Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual, 2d Ed, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., Ch. 18, pp. 64-75. An HIV-1 positive serum diluted 1000-fold, or a human Fab derived from repertoire cloning was used to react with the fusion proteins. The second antibody was HRP-conjugated goat antihuman Fab. An ECL Western blotting detection system (Amersham) was used to detect the bound antibody. A detailed protocol for this detection system was provided by the manufacturer. Rainbow molecular weight markers (Amersham) were used to estimate the size of fusion proteins.
  • CEM cells fusion assays were performed as previously described (Matthews et al., 1987, Proc. Natl. Acad. Sci. USA 84: 5424-5481). CEM cells (7 ⁇ 10 4 ) were incubated with HIV-1 IIIB chronically infected CEM cells (10 4 ) in 96-well flat-bottomed half-area plates (Costar) in 100 culture medium. Peptide and fusion proteins at various concentrations in 10 ⁇ l culture medium were incubated with the cell mixtures at 37° C. for 24 hours. Multinucleated syncytia were estimated with microscopic examination. Both M41 and M41-P did not show cytotoxicity at the concentrations tested and shown in FIG. 8 .
  • Inhibition of HIV-1 induced cell-cell fusion activity was carried out in the presence of 10 nM DP178 and various concentrations of M41 ⁇ 178 or M41-P ⁇ 178 as indicated in FIG. 9 . There was no observable syncytia in the presence of 10 nM DP178. No peptide or fusion protein was added in the control samples.
  • the amino acid sequence of DP178 used is: YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF(SEQ ID NO:1).
  • ELISA enzyme linked immunoassay
  • M41 ⁇ 178 or M41-P ⁇ 178 (5 ⁇ g/ml) in 0.1M NaHCO 3 , pH 8.6, were coated on 96 wells Linbro ELISA plates (Flow Lab, Inc.) overnight. Each well was washed three times with distilled water then blocked with 3% bovine serum albumin (BSA) for 2 hours.
  • BSA bovine serum albumin
  • peptides with 0.5% BSA in TBST 40 mM Tris-HCl pH7.5, 150 mM NaCl, 0.05% Tween 20 were added to the ELISA plates and incubated at room temperature for 1 hour.
  • Fab-d was added at a concentration of 10 ng/ml with 0.5% BSA in TBST. The plates were washed three times with TBST after incubation at room temperature for 1 hour.
  • HRP horse radish peroxidase
  • the plates were then washed four times with TBST.
  • the peroxidase substrate o-phenylene diamine (2.5 mg/ml) and 0.15% H 2 O 2 were added to develop the color.
  • the reaction was stopped with an equal volume of 4.5 NH 2 SO 4 after incubation at room temperature for 10 minutes.
  • the optical density of the stopped reaction mixture was measured with a micro plate reader (Molecular Design) at 490 nm. Results are shown in FIG. 10 .
  • the ectodomain of gp41 was expressed as a maltose binding fusion protein (M41) ( FIG. 7 ).
  • M41 maltose binding fusion protein
  • the fusogenic peptide sequence at the N-terminal of gp41 was omitted from this recombinant protein and its derivatives to improve solubility.
  • the maltose binding protein facilitated purification of the fusion proteins under relatively mild, non-denaturing conditions.
  • M41 soluble recombinant gp41 was not glycosylated, lacked several regions of the transmembrane protein (i.e., the fusion peptide, the membrane spanning, and the cytoplasmic domains), and was expressed in the absence of gp120, it was not expected to precisely reflect the structure of native gp41 on HIV-1 virions. Nevertheless, purified M41 folded in a manner that preserved certain discontinuous epitopes as evidenced by reactivity with human monoclonal antibodies, 98-6, 126-6, and 50-69, previously shown to bind conformational epitopes on native gp41 expressed in eukaryotic cells (Xu et al., 1991, J. Virol.
  • the wild type M41 fusion protein was tested for anti-HIV-1 activity.
  • synthetic peptides corresponding to the leucine zipper (DP107) and the C-terminal putative helix (DP178) show potent anti-HIV activity.
  • the recombinant M41 protein did not affect HIV-1 induced membrane fusion at concentrations as high as 50 ⁇ M (Table XXV, below).
  • RT activity was determined by a modification of the published methods of Goff et al., 1981, J. Virol. 38: 239-248 and Willey et al., 1988, J. Virol. 62: 139-147 as described in Chen et al., 1993, AIDS Res. Human Retroviruses 9: 1079-1086.
  • M41-P fusion protein
  • Table XXV and FIG. 8 a single amino acid substitution, proline in place of isoleucine in the middle of the leucine zipper motif, yielded a fusion protein (M41-P) which did exhibit antiviral activity (Table XXV and FIG. 8 ).
  • M41-P blocked syncytia formation by 90% at approximately 85 nM and neutralized HIV-1 IIIB infection by 90% at approximately 70 nM concentrations.
  • the anti-HIV-1 activity of M41-P appeared to be mediated by the C-terminal helical sequence since deletion of that region from M41-P yielded an inactive fusion protein, M41-P ⁇ 178 (Table XXV).
  • Fab-d failed to bind either the DP178 peptide or the fusion protein M41 ⁇ 178, but its epitope was reconstituted by simply mixing these two reagents together ( FIG. 10 ).
  • the proline mutation in the leucine zipper domain of the fusion protein, M41-P ⁇ 178 failed to reconstitute the epitope in similar mixing experiments.
  • Coiled-coil nomenclature labels each of seven amino acids of a heptad repeat A through G, with amino acids A and D tending to be hydrophobic positions. Amino acids E and G tend to be charged. These four positions (A, D, E, and G) form the amphipathic backbone structure of a monomeric alpha-helix. The backbones of two or more amphipathic helices interact with each other to form di-, tri-, tetrameric, etc., coiled-coil structures.
  • coiled coils including yeast transcription factor GCN4, Influenza Virus hemagglutinin loop 36, and human proto-oncogenes c-Myc, c-Fos, and c-Jun.
  • yeast transcription factor GCN4 Influenza Virus hemagglutinin loop 36
  • human proto-oncogenes c-Myc, c-Fos, and c-Jun For each peptide sequence, a strict homology for the A and D positions, and a list of the amino acids which could be excluded for the B, C, E, F, and G positions (because they are not observed in these positions) was determined.
  • Motifs were tailored to the DP107 and DP178 sequences by deducing the most likely possibilities for heptad positioning of the amino acids of HIV-1 Bru DP-107, which is known to have coiled-coil structure, and HIV-1 Bru DP178, which is still structurally undefined.
  • the analysis of each of the sequences is contained in FIG. 12 .
  • the motif for GCN4 was designed as follows:
  • FIG. 13 illustrates several possible sequence alignments for both DP107 and DP178 and also includes motif designs based on 28-mer, 35-mer, and full-length peptides. Notice that only slight differences occur in the motifs as the peptides are lengthened. Generally, lengthening the base peptide results in a less stringent motif. This is very useful in broadening the possibilities for identifying DP107- or DP-178-like primary amino acid sequences referred to in this document as “hits”.
  • hybrid motifs are made by “crossing” two or more very stringent motifs to make a new search algorithm which will find not only both “parent” motif sequences but also any peptide sequences which have similarities to one, the other, or both “parents”. For example, in FIG. 14 the “parent” sequence of GCN4 is crossed with each of the possible “parent” motifs of DP-107. Now the hybrid motif must contain all of the amino acids found in the A and D positions of both parents, and exclude all of the amino acids not found in either parent at the other positions.
  • Hybridizations can be performed on any combination of two or more motifs.
  • FIG. 17 summarizes several three-motif hybridizations including GCN4, DP107 (both frames), and DP178 (also both frames). Notice that the resulting motifs are now becoming much more similar to each other. In fact, the first and third hybrid motifs are actually subsets of the second and fourth hybrid motifs respectively. This means that the first and third hybrid motifs are slightly more stringent than the second and fourth. It should also be noted that with only minor changes in these four motifs, or by hybridizing them, a single motif could be obtained which would find all of the sequences. However, it should be remembered that stringency is also reduced. Finally, the most broad-spectrum and least-stringent hybrid motif is described in FIG. 18 which summarizes the hybridization of GCN4, DP107 (both frames), DP178 (both frames), c-Fos, c-Jun, c-Myc, and Flu loop 36.
  • DP-178 is located only approximately ten amino acids upstream of the transmembrane spanning region of gp41 and just C-terminal to a proline which separates DP107 and DP178. It has been postulated that DP178 may be an amphipathic helix when membrane associated, and that the proline might aid in the initiation of the helix formation. The same arrangement was observed in Respiratory Syncytial Virus; however, the DP178-like region in this virus also had a leucine zipper just C-terminal to the proline. Therefore, N-terminal proline-leucine zipper motifs were designed to analyze whether any other viruses might contain this same pattern. The motifs are summarized in FIG. 19 .
  • the PC/Gene protein database contains 5879 viral amino acid sequences (library file PVIRUSES; CD-ROM release 11.0). Of these, 1092 are viral enveloped or glycoprotein sequences (library file PVIRUSE1). Tables V through XIV contain lists of protein sequence names and motif hit locations for all the motifs searched.
  • FIG. 20 represents search results for HIV-1 BRU isolate gp41 (PC/Gene protein sequence PENV_HV1BR). Notice that the hybrid motif which crosses DP-107 and DP-178 (named 107 ⁇ 178 ⁇ 4; the same motif as found in FIG. 16 found three hits including amino acids 550-599, 636-688, and 796-823. These areas include DP-107 plus eight N-terminal and four C-terminal amino acids; DP178 plus seven N-terminal and ten C-terminal amino acids; and an area inside the transmembrane region (cytoplasmic). FIG. 20 also contains the results obtained from searching with the motif named ALLMOTI5, for which the key is found in FIG. 17 ( ⁇ CDGHP ⁇ ⁇ CFP ⁇ 5).
  • ALLMOTI5 for which the key is found in FIG. 17 ( ⁇ CDGHP ⁇ ⁇ CFP ⁇ 5).
  • This motif also found three hits including DP107 (amino acids 510-599), DP178 (615-717), and a cytoplasmic region (772-841). These hits overlap the hits found by the motif 107 ⁇ 178 ⁇ 4 with considerable additional sequences on both the amino and carboxy termini. This is not surprising in that 107 ⁇ 178 ⁇ 4 is a subset of the ALLMOTI5 hybrid motif. Importantly, even though the stringency of ALLMOTI5 is considerably less than 107 ⁇ 178 ⁇ 4, it still selectively identifies the DP107 and DP178 regions of gp41 shown to contain sequences for inhibitory peptides of HIV-1. The results of these two motif searches are summarized in Table V under the PC/Gene protein sequence name PENV_HV1BR.
  • proline-leucine zipper motifs also gave several hits in HIV-1 BRU including 503-525 which is at the very C-terminus of gp120, just upstream of the cleavage site (P7LZIPC and P12LZIPC); and 735-768 in the cytoplasmic domain of gp41 (P23LZIPC). These results are found in Tables VIII, IX, and X under the same sequence name as mentioned above. Notice that the only area of HIV-1 BRU which is predicted by the Lupas algorithm to contain a coiled-coil region, is from amino acids 635-670. This begins eight amino acids N-terminal to the start and ends eight amino acids N-terminal to the end of DP178. DP107, despite the fact that it is a known coiled coil, is not predicted to contain a coiled-coil region using the Lupas method.
  • FIG. 21 represents search results for Human Respiratory Syncytial Virus (RSV; Strain A2) fusion glycoprotein F1 (PC/Gene protein sequence name PVGLF_HRSVA).
  • Motif 107 ⁇ 178 ⁇ 4 finds three hits including amino acids 152-202, 213-243, and 488-515. The arrangement of these hits is similar to what is found in HIV-1 except that the motif finds two regions with similarities to DP-178, one just downstream of what would be called the DP107 region or amino acids 213-243, and one just upstream of the transmembrane region (also similar to DP178) or amino acids 488-515.
  • Motif ALLMOTI5 also finds three areas including amino acids 116-202, 267-302, and 506-549.
  • proline-leucine zipper motifs also gave several hits including amino acids 205-221 and 265-287 (P1LZIPC 265-280, P12LZIPC), and 484-513 (P7LZIPC and P12LZIPC 484-506, P23LZIPC). Notice that the PLZIP motifs also identify regions which share location similarities with DP-178 of HIV-1.
  • Motif hits for Simian immunodeficiency Virus gp41 are shown in FIG. 22 .
  • Motif 107 ⁇ 178 ⁇ 4 finds three hits including amino acids 566-593, 597-624, and 703-730. The first two hits only have three amino acids between them and could probably be combined into one hit from 566-624 which would represent a DP107-like hit. Amino acids 703 to 730 would then represent a DP178-like hit.
  • ALLMOTI5 also finds three hits including amino acids 556-628 (DP107-like), 651-699 (DP178-like), and 808-852 which represents the transmembrane spanning region.
  • SIV also has one region from 655-692 with a high propensity to form a coiled coil as predicted by the Lupas algorithm. Both 107 ⁇ 178 ⁇ 4 and ALLMOTI5 motifs find the same region. SIV does not have any PLZIP motif hits in gp41.
  • Canine Distemper Virus (strain Onderstepoort) fusion glycoprotein F1 (PC/Gene Protein sequence name PVGLF_CDVO) has regions similar to Human RSV which are predicted to be DP107-like and DP178-like ( FIG. 23 ). Motif 107 ⁇ 178 ⁇ 4 highlights one area just C-terminal to the fusion peptide at amino acids 252-293. Amino acids 252-286 are also predicted to be coiled coil using the Lupas algorithm. Almost 100 amino acids C-terminal to the first region is a DP178-like area at residues 340-367.
  • ALLMOTI5 highlights three areas of interest including: amino acids 228-297, which completely overlaps both the Lupas prediction and the DP107-like 107 ⁇ 178 ⁇ 4 hit; residues 340-381, which overlaps the second 107 ⁇ 178 ⁇ 4 hit; and amino acids 568-602, which is DP178-like in that it is located just N-terminal to the transmembrane region. It also overlaps another region (residues 570-602) predicted by the Lupas method to have a high propensity to form a coiled coil.
  • FIG. 24 shows the motif hits found in Newcastle Disease Virus (strain Australia-Victoria/32; PC Gene protein sequence name PVGLF_NDVA).
  • Motif 107 ⁇ 178 ⁇ 4 finds two areas including a DP107-like hit at amino acids 151-178 and a DP178-like hit at residues 426-512.
  • ALLMOTI5 finds three areas including residues 117-182, 231-272, and 426-512.
  • the hits from 426-512 include a region which is predicted by the Lupas method to have a high coiled-coil propensity (460-503).
  • the PLZIP motifs identify only one region of interest at amino acids 273-289 (P1 and 12LZIPC).
  • Both motifs 107 ⁇ 178 ⁇ 4 and ALLMOTI5 exhibit DP107-like hits in the same region, 115-182 and 117-182 respectively, of Human Parainfluenza Virus (strain NIH 47885; PC/Gene protein sequence name PVGLF_p13H4; ( FIG. 25 ).
  • the two motifs have a DP178-like hit just slightly C-terminal at amino acids 207-241. Both motifs also have DP178-like hits nearer the transmembrane region including amino acids 457-497 and 462-512 respectively.
  • PLZIP motif hits are also observed including 283-303 (P5LZIPC), 283-310 (P12LZIPC), 453-474 (P6LZIPC), and 453-481 (P23LZIPC).
  • the Lupas algorithm predicts that amino acids 122-176 may have a propensity to form a coiled-coil.
  • FIG. 26 illustrates the Lupas prediction for a coiled coil in Influenza A Virus (strain A/Aichi/2/68) at residues 379-436, as well as the motif hits for 107 ⁇ 178 ⁇ 4 at amino acids 387-453, and for ALLMOTI5 at residues 380-456.
  • Residues 383-471 38-125 of HA2 were shown by Carr and Kim to be an extended coiled coil when under acidic pH (Carr and Kim, 1993, Cell 73: 823-832).
  • the Lupas algorithm predicts a coiled-coil at residues 379-436. All three methods successfully predicted the region shown to actually have coiled-coil structure; however, ALLMOTI5 predicted the greatest portion of the 88 residue stretch.
  • respiratory syncytial virus (RSV) peptides identified by utilizing the computer-assisted search motifs described in the Examples presented in Sections 9 and 11, above, were tested for anti-RSV activity. Additionally, circular dichroism (CD) structural analyses were conducted on the peptides, as discussed below. It is demonstrated that several of the identified peptides exhibit potent antiviral capability. Additionally, it is shown that several of these peptides exhibit a substantial helical character.
  • RSV respiratory syncytial virus
  • the CD spectra were measured in a 10 mM sodium phosphate, 150 mM sodium chloride, pH 7.0, buffer at approximately 10 mM concentrations, using a 1 cm pathlength cell on a Jobin/Yvon Autodichrograph Mark V CD spectrophotometer.
  • Peptides were synthesized according to the methods described, above, in Section 6.1. Peptide concentrations were determined from A 280 using Edleroch's method (1967, Biochemistry 6:1948).
  • Anti-RSV antiviral activity assays The assay utilized herein tested the ability of the peptides to disrupt the ability of HEp2 cells acutely infected with RSV (i.e., cells which are infected with a multiplicity of infection of greater than 2) to fuse and cause syncytial formation on a monolayer of uninfected an uninfected line of Hep-2 cells. The lower the observed level of fusion, the greater the antiviral activity of the peptide was determined to be.
  • Uninfected confluent monolayers of Hep-2 cells were grown in microtiter wells in 3% EMEM (Eagle Minimum Essential Medium w/o L-glutamine [Bio Whittaker Cat. No. 12-125F], with fetal bovine serum [FBS; which had been heat inactivated for 30 minutes at 56° C.; Bio Whittaker Cat. No. 14-501F) supplemented at 3%, antibiotics (penicillin/streptomycin; Bio Whittaker Cat. No. 17-602E) added at 1%, and glutamine added at 1%.
  • EMEM Eagle Minimum Essential Medium w/o L-glutamine [Bio Whittaker Cat. No. 12-125F]
  • FBS fetal bovine serum
  • antibiotics penicillin/streptomycin
  • Bio Whittaker Cat. No. 17-602E glutamine added at 1%.
  • Hep2 cells for addition to uninfected cells, cultures of acutely infected Hep2 cells were washed with DPBS (Dulbecco's Phosphate Buffered Saline w/o calcium or magnesium; Bio Whittaker Cat. No. 17-512F) and cell monolayers were removed with Versene (1:5000; Gibco Life Technologies Cat. No. 15040-017). The cells were spun 10 minutes and resuspended in 3% FBS. Cell counts were performed using a hemacytometer. Persistent cells were added to the uninfected Hep-2 cells.
  • DPBS Dynabecco's Phosphate Buffered Saline w/o calcium or magnesium
  • Versene (1:5000; Gibco Life Technologies Cat. No. 15040-017
  • the cells were spun 10 minutes and resuspended in 3% FBS. Cell counts were performed using a hemacytometer. Persistent cells were added to the uninfected Hep-2 cells.
  • the antiviral assay was conducted by, first, removing all media from the wells containing uninfected Hep-2 cells, then adding peptides (at the dilutions described below) in 3% EMEM, and 100 acutely RSV-infected Hep2 cells per well. Wells were then incubated at 37° C. for 48 hours.
  • Crystal Violet stain approximately 50 ⁇ l 0.25% Crystal Violet stain in methanol were added to each well. The wells were rinsed immediately, to remove excess stain, and were allowed to dry. The number of syncytia per well were then counted, using a dissecting microscope.
  • XTT 2,3-bis[2-Methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxyanilide inner salt
  • 50 ⁇ l XTT (1 mg/ml in RPMI buffered with 100 mM HEPES, pH 7.2-7.4, plus 5% DMSO) were added to each well.
  • the OD 450/690 was measured (after blanking against growth medium without cells or reagents, and against reagents) according to standard procedures.
  • Peptides The peptides characterized in the study presented herein were:
  • the peptides of group 1 represent portions of the RSV F2 protein DP178/107-like region.
  • the peptides of group 2 represent portions of the RSV F1 protein DP107-like region.
  • the peptides of groups 3 represent portions of the RSV F1 protein DP178-like region.
  • Each peptide was tested at 2-fold serial dilutions ranging from 100 ⁇ g/ml to approximately 100 ng/ml. For each of the assays, a well containing no peptide was also used. The IC 50 data for each peptide represents the average of several experiments conducted utilizing that peptide.
  • FIGS. 27A-C and 28 A-C represent antiviral and structural information obtained from peptides derived from the RSV F2 DP178/DP107-like F2 region ( FIG. 27A-C ), the RSV F1 DP-107-like region ( FIG. 27D-F ) and the RSV DP178-like F2 region ( FIG. 28A-C ).
  • FIGS. 27A-F a number of the RSV DP178/DP107-like peptides exhibited a detectable level of antiviral activity.
  • Peptides from the RSV DP178/DP107-like F2 region for example, T-142 to T-145 and T-334 purfied peptides, exhibited detectable levels of antiviral activity, as evidenced by their IC 50 values.
  • a number of RSV F1 DP107-like peptides FIG.
  • FIG. 27D-F exhibited a sizable level of antiviral activity as purified peptides, including, for example, peptides T-124 to T-127, T-131, T-135 and T-137 to T-139, as demonstrated by their low IC 50 values.
  • CD analysis FIG. 27A-B , 27 D-E) reveals that many of the peptides exhibit some detectable level of helical structure.
  • FIG. 28A-C demonstrate that a number of DP178-like purified peptides exhibit a range of potent anti-viral activity. These peptides include, for example, T-67, T-104, T-105 and T-107 to T-119, as listed in FIG. 28A-B , and T-665 to 1-669 and T-671 to 1-673, as listed in FIG. 28C . In addition, some of the DP178-like peptides exhibited some level of helicity.
  • HPIV3 human parainfluenza virus type 3 peptides identified by utilizing the computer-assisted search motifs described in the Examples presented in Sections 9 and 15, above, were tested for anti-HPIV3 activity. Additionally, circular dichroism (CD) structural analyses were conducted on the peptides, as discussed below. It is demonstrated that several of the identified peptides exhibit potent antiviral capability. Additionally, it is shown that several of these peptides exhibit a substantial helical character.
  • CD circular dichroism
  • CD circular dichroism
  • Anti-HPIV3 antiviral activity assays The assay utilized herein tested the ability of the peptides to disrupt the ability of Hep2 cells chronically infected with HPIV3 to fuse and cause syncytial formation on a monolayer of an uninfected line of CV-1W cells. The more potent the lower the observed level of fusion, the greater the antiviral activity of the peptide.
  • Hep2 cells for addition to uninfected cells, cultures of chronically infected Hep2 cells were washed with DPBS (Dulbecco's Phosphate Buffered Saline w/o calcium or magnesium; Bio Whittaker Cat. No. 17-512F) and cell monolayers were removed with Versene (1:5000; Gibco Life Technologies Cat. No. 15040-017). The cells were spun 10 minutes and resuspended in 3% FBS. Cell counts were performed using a hemacytometer. Persistent cells were added to the uninfected CV-1W cells.
  • DPBS Dynabecco's Phosphate Buffered Saline w/o calcium or magnesium
  • Versene (1:5000; Gibco Life Technologies Cat. No. 15040-017
  • the cells were spun 10 minutes and resuspended in 3% FBS. Cell counts were performed using a hemacytometer. Persistent cells were added to the uninfected CV-1W cells.
  • the antiviral assay was conducted by, first, removing all media from the wells containing uninfected CV-1W cells, then adding peptides (at the dilutions described below) in 3% EMEM, and 500 chronically HPIV3-infected Hep2 cells per well. Wells were then incubated at 37° C. for 24 hours.
  • Peptides The peptides characterized in the study presented herein were:
  • Each peptide was tested at 2-fold serial dilutions ranging from 500 ⁇ g/ml to approximately 500 ng/ml. For each of the assays, a well containing no peptide was also used.
  • FIGS. 29A-E and 30 A-C represent antiviral and structural information obtained from peptides derived from the HPIV3 fusion protein DP107-like region ( FIG. 29A-E ) and the HPIV3 fusion protein DP178-like region ( FIG. 30A-C ).
  • HPIV3 DP107-like peptides exhibited potent levels of antiviral activity.
  • These peptides include, for example, peptides T-40, T-172 to T-175, T-178, T-184 and T-185.
  • CD analysis reveals that a number of the peptides exhibit detectable to substantial level of helical structure.
  • results summarized in FIG. 30A-C demonstrate that a number of the DP178-like peptides tested exhibit a range of anti-viral activity. These peptides include, for example, peptides 194 to 211, as evidenced by their low IC 50 values. In fact, peptides 201 to 205 exhibit IC 50 values in the nanogram/ml range. In addition, many of the DP178-like peptides exhibited some level of helicity.
  • FIG. 31 represents search results for SIV isolate MM251 (PC/Gene® protein sequence PENV_SIVM2). Both 107 ⁇ 178 ⁇ 4 and ALLMOTI5 search motifs identified two regions with similarities to DP107 and/or DP178.
  • the peptide regions found by 107 ⁇ 178 ⁇ 4 were located at amino acid residues 156-215 and 277-289.
  • the peptide regions found by ALLMOTI5 were located at amino acid residues 156-219 and 245-286. Both motifs, therefore, identify similar regions.
  • the first SIV peptide region correlates with a DP107 region
  • the second region identified correlates with the DP178 region of HIV.
  • an alignment of SIV isolate MM251 and HIV isolate BRU, followed by a selection of the best peptide matches for HIV DP107 and DP178, reveals that the best matches are found within the peptide regions identified by the 107 ⁇ 178 ⁇ 4 and ALLMOTI5 search motifs.
  • a potential coiled-coil region at amino acid residues 242-282 is predicted by the Lupas program. This is similar to the observation in HIV in which the coiled-coil is predicted by the Lupas program to be in the DP178 rather than in the DP107 region. It is possible, therefore, that SIV may be similar to HIV in that it may contain a coiled-coil structure in the DP107 region, despite such a structure being missed by the Lupas algorithm. Likewise, it may be that the region corresponding to a DP178 analog in SIV may exhibit an undefined structure, despite the Lupas program's prediction of a coiled-coil structure.
  • Epstein-Barr is a human herpes virus which is the causative agent of, for example, infectious mononucleosis (IM), and is also associated with nasopharyngeal carcinomas (NPC), Burkitt's lymphoma and other diseases.
  • IM infectious mononucleosis
  • NPC nasopharyngeal carcinomas
  • Burkitt's lymphoma Burkitt's lymphoma and other diseases.
  • the virus predominantly exists in the latent form and is activated by a variety of stimuli.
  • FIG. 32 depicts the search motif results for the Epstein-Barr Virus (Strain B95-8; PC/Gene® protein sequence PVGLB_EBV) glycoprotein gp110 precursor (gp115).
  • the 107 ⁇ 178 ⁇ 4 motif identified two regions of interest, namely the regions covered by amino acid residues 95-122 and 631-658.
  • One PZIP region was identified at amino acid residue 732-752 which is most likely a cytoplasmic region of the protein.
  • the Lupas algorithm predicts a coiled-coil structure for amino acids 657-684. No ALLMOTI5 regions were identified.
  • FIG. 33 depicts the search motif results for the Zebra (or EB1) trans-activator protein (BZLF1) of the above-identified Epstein-Barr virus.
  • This protein is a transcription factor which represents the primary mediator of viral reactivation. It is a member of the b-ZIP family of transcription factors and shares significant homology with the basic DNA-binding and dimerization domains of the cellular oncogenes c-fos and C/EBP.
  • the Zebra protein functions as a homodimer.
  • Search results domonstrate that the Zebra protein exhibits a single region which is predicted to be either of DP107 or DP178 similarity, and is found between the known DNA binding and dimerization regions of the protein. Specifically, this region is located at amino acid residues 193-220, as shown in FIG. 33 .
  • the Lupas program predicted no coiled-coil regions.
  • FIG. 34 illustrates the motif search results for the fusion protein F1 of measles virus, strain Edmonston (PC Gene® protein sequence PVGLF MEASE), successfully identifying DP178/DP107 analogs.
  • the 107 ⁇ 178 ⁇ 4 motif identifies a single region at amino acid residues 228-262.
  • the ALLMOTI5 search motif identifies three regions, including amino acid residues 116-184, 228-269 and 452-500. Three regions containing proline residues followed by a leucine zipper-like sequence were found beginning at proline residues 214, 286 and 451.
  • the Lupas program identified two regions it predicted had potential for coiled-coil structure, which include amino acid residues 141-172 and 444-483.
  • FIG. 35 depicts the results of a PZIP motif search conducted on the Hepatitis B virus subtype AYW. Two regions of interest within the major surface antigen precursor S protein were identified. The first lies just C-terminal to the proposed fusion peptide of the major surface antigen (Hbs) which is found at amino acid residues 174-191. The second region is located at amino acid residues 233-267. The Lupas program predicts no coiled-coil repeat regions.
  • peptides derived from area around the analog regions are synthesized, as shown in FIG. 52A-B . These peptides represent one amino acid peptide “walks” through the putative DP178/DP107 analog regions.
  • the peptides are synthesized according to standard Fmoc chemistry on Rinkamide MBHA resins to provide for carboxy terminal blockade (Chang, C. D. and Meinhofer, J., 1978, Int. J. Pept. Protein Res. 11:246-249; Fields, G. B. and Noble, R. L., 1990, Int. J. Pept. Protein Res. 35:161-214).
  • the anti-HBV activity of the peptides is tested by utilizing standard assays to determine the test peptide concentration required to cause an acceptable (e.g., 90%) decrease in the amount of viral progeny formed by cells exposed to an HBV viral inoculum.
  • Candidate antivial peptides are further characterized in model systems such as wood chuck tissue culture and animal sytems, prior to testing on humans.
  • the results depicted herein illustrate the results of search motifs conducted on the Simian Mason-Pfizer monkey virus.
  • the motifs reveal DP178/DP107 analogs within the enveloped (TM) protein GP20, as shown in FIG. 36 .
  • the 107 ⁇ 178 ⁇ 4 motifs identifies a region at amino acid residues 422-470.
  • the ALLMOTI5 finds a region at amino acid residues 408-474.
  • the Lupas program predicted a coiled-coil structure a amino acids 424-459.
  • FIG. 37 depicts the search motif results for the Pseudomonas aeruginosa fimbrial protein (Pilin). Two regions were identified by motifs 107 ⁇ 178 ⁇ 4 and ALLMOTI5. The regions located at amino acid residues 30-67 and 80-144 were identified by the 107 ⁇ 178 ⁇ 4 motif. The regions at amino acid residues 30-68 and 80-125 were identified by the ALLMOTI5.
  • FIG. 38 depicts the search motif results for the Pseudomonas gonorrhoeae fimbrial protein (Pilin).
  • Pilin Pseudomonas gonorrhoeae fimbrial protein
  • FIG. 39 depicts the search motif results for the Hemophilus Influenza fimbrial protein (Pilin).
  • a single region was identified by both the 107 ⁇ 178 ⁇ 4 and the ALLMOTI5 motifs.
  • the region located at amino acid residues 102-129 was identified by the 107 ⁇ 178 ⁇ 4 motif.
  • the region located at amino acid residues 102-148 were identified by the ALLMOTI5 search motif. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 40 depicts the search motif results for the Staphylococcus aureus toxic shock syndrome Hemophilus Influenza fimbrial protein (Pilin).
  • a single region was identified by both the 107 ⁇ 178 ⁇ 4 and the ALLMOTI5 motifs.
  • the region located at amino acid residues 102-129 was identified by the 107 ⁇ 178 ⁇ 4 motif.
  • the region located at amino acid residues 102-148 were identified by the ALLMOTI5 search motif. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 41 summarizes the motif search results conducted on the Staphylococcus aureus enterotoxin Type E protein. These results demonstrate the successful identification of DP178/DP107 analogs corresponding to peptide sequences within this protein, as described below.
  • the ALLMOTI5 motif identified a region at amino acid residues 22-27.
  • the 107 ⁇ 178 ⁇ 4 motif identified two regions, with the first at amino acid residues 26-69 and the second at 88-115.
  • a P12LZIPC motif search identified two regions, at amino acid residues 163-181 and 230-250.
  • the Lupas program predicted a region with a high propensity for coiling at amino acid residues 25-54. This sequence is completely contained within the first region identified by both ALLMOTI5 and 107 ⁇ 178 ⁇ 4 motifs.
  • FIG. 42 depicts the search motif results conducted on a second Staphylococcus aureus toxin, enterotoxin A. Two regions were identified by the ALLMOTI5 motif, at amino acid residues 22-70 and amino acid residues 164-205. The 107 ⁇ 178 ⁇ 4 motif found two regions, the first at amino acid residues 26-69 and the second at amino acid residues 165-192. A P23LZIPC motif search revealed a region at amino acid residues 216-250. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 43 shows the motif search results conducted on the E. coli heat labile enterotoxin A protein, demonstrating that identification of DP178/DP107 analogs corresponding to peptides located within this protein.
  • Two regions were identified by the ALLMOTI5 motif, with the first residing at amino acid residues 55-115, and the second residing at amino acid residues 216-254.
  • the 107 ⁇ 178 ⁇ 4 motif identified a single region at amino acid residues 78-105. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 44 illustrates the search motif results conducted on the human c-fos oncoprotein.
  • the ALLMOTI5 motif identified a single region at amino acid residues 155-193.
  • the 107 ⁇ 178 ⁇ 4 motif identified one region at amino acid residues 162-193.
  • the Lupas program predicted a region at amino acid residues 148-201 to have coiled-coil structure.
  • FIG. 45 illustrates the search motif results conducted on the human lupus KU autoantigen protein P70.
  • the ALLMOTI5 motif identified a single region at amino acid residues 229-280.
  • the 107 ⁇ 178 ⁇ 4 motif identified one region at amino acid residues 235-292.
  • the Lupas program predicted a region at amino acid residues 232-267 to have coiled-coil structure.
  • FIG. 46 illustrates the search motif results conducted on the human zinc finger protein 10.
  • ALLMOTI5 motif identified a single region at amino acid residues 29-81.
  • the 107 ⁇ 178 ⁇ 4 motif identified one region at amino acid residues 29-56.
  • a P23LZIPC motif search found a single region at amino acid residues 420-457.
  • the Lupas program predicted no coiled-coil regions.
  • CD circular dichroism
  • the CD spectra were measured in a 10 mM sodium phosphate, 150 mM sodium chloride, pH 7.0, buffer at approximately 10 mM concentrations, using a 1 cm pathlength cell on a Jobin/Yvon Autodichrograph Mark V CD spectrophotometer. Peptide concentrations were determined from A 280 using Edleroch's method (1967, Biochemistry 6:1948).
  • Anti-MeV antiviral activity syncytial reduction assay The assay utilized herein tested the ability of the peptides to disrupt the ability of Vero cells acutely infected with MeV (i.e., cells which are infected with a multiplicity of infection of 2-3) to fuse and cause syncytial formation on a monolayer of an uninfected line of Vero cells. The more potent the peptide, the lower the observed level of fusion, the greater the antiviral activity of the peptide.
  • the antiviral assay was conducted by, first, removing all media from the wells containing uninfected Vero cells, then adding peptides (at the dilutions described below) in 10% FBS EMEM, and 50-100 acutely MeV-infected Vero cells per well. Wells were then incubated at 37° C. for a maximum of 18 hours.
  • Anti-MeV antiviral activity plaque reduction assay The assay utilized herein tested the ability of the peptides to disrupt the ability of MeV to infect permissive, uninfected Vero cells, leading to the infected cells' fusing with uninfected cells to produce syncytia. The lower the observed level of syncytial formation, the greater the antiviral activity of the peptide.
  • Monolayers of uninfected Vero cells are grown as described above.
  • the antiviral assay was conducted by, first, removing all media from the wells containing uninfected Vero cells, then adding peptides (at the dilutions described below) in 10% FBS EMEM, and MeV stock virus at a final concentration of 30 plaque forming units (PFU) per well. Wells were then incubated at 37° C. for a minimum of 36 hours and a maximum of 48 hours.
  • Peptides The peptides characterized in the study presented herein were peptides T-252A0 to T-256A0, T-25781/C1, and T-258B1 to T-265B0, and T-266A0 to T-268A0, as shown in FIG. 47A-B . These peptides represent a walk through the DP178-like region of the MeV fusion protein.
  • Each peptide was tested at 2-fold serial dilutions ranging from 100 ⁇ g/ml to approximately 100 ng/ml. For each of the assays, a well containing no peptide was also used.
  • FIG. 47A-B The data summarized in FIG. 47A-B represents antiviral and structural information obtained via “peptide walks” through the DP178-like region of the MeV fusion protein.
  • the MeV DP178-like peptides exhibited a range of antiviral activity as crude peptides. Several of these peptides were chosen for purification and further antiviral characterization. The IC 50 values for such peptides were determined, as shown in FIG. 47A-B , and ranged from 1.35 ⁇ g/ml (T-257B1/C1) to 0.072 ⁇ g/ml (T-265B1). None of the DP178-like peptides showed, by CD analysis, a detectable level of helicity.
  • SIV simian immunodeficiency virus
  • Peptides The peptides characterized in the study presented herein were peptides T-391 to T-400, as shown in FIG. 48A-B . These peptides represent a walk through the DP178-like region of the SIV TM protein.
  • Each peptide was tested at 2-fold serial dilutions ranging from 100 ⁇ g/ml to approximately 100 ng/ml. For each of the assays, a well containing no peptide was also used.
  • the data summarized in FIG. 48A-B represents antiviral information obtained via “peptide walks” through the DP178-like region of the SIV TM protein.
  • peptides T-391 to T-400 were tested and exhibited a potent antiviral activity as crude peptides.
  • Example presented in this Section represents a study of the antiviral activity of DP107 and DP178 truncations and mutations. It is demonstrated that several of these DP107 and DP178 modified peptides exhibit substantial antiviral activity.
  • Anti-HIV assays The antiviral assays performed were as those described, above, in Section 6.1. Assays utilized HIV-1/IIIb and/or HIV-2 NIHZ isolates. Purified peptides were used, unless otherwise noted in FIGS. 49A-L .
  • Peptides The peptides characterized in the study presented herein were:
  • Anti-HIV antiviral data was obtained with the group 1 DP178-derived peptides listed in FIG. 49A-L .
  • the full-length, non-mutant DP178 peptide (referred to in FIG. 49A-L as T20) results shown are for 4 ng/ml.
  • a number of the DP178 truncations exhibited a high level of antiviral activity, as evidenced by their low IC 50 values.
  • These include, for example, test peptides T-50, T-624, T-636 to T-641, T-645 to T-650, T-652 to T-654 and T-656.
  • T-50 represents a test peptide which contains a point mutation, as indicated by the residue's shaded background.
  • the HIV-1-derived test peptides exhibited a distinct strain-specific antiviral activity, in that none of the peptides tested on the HIV-2 NIHZ isolate demonstrated appreciable antti-HIV-2 antiviral activity.
  • test peptides representing the amino (T-4) and carboxy (T-3) terminal halves of DP178 were tested.
  • IC 50 3 ⁇ g/ml
  • a number of additional test peptides also exhibited a high level of antiviral activity.
  • T-61/T-102 T-217 to T-221, T-235, T-381, T-677, T-377, T-590, T-378, T-591, T-271 to T-272, T-611, T-222 to T-223 and T-60/T-224.
  • Certain of the antiviral peptides contain point mutations and/or amino acid residue additions which vary from the DP178 amino acid sequence.
  • FIG. 49I-L point mutations and/or amino and/or carboxy-terminal modifications are introduced into the DP178 amino acid sequence itself. As shown in the figure, the majority of the test peptides listed exhibit potent antiviral activity.
  • Truncations of the DP107 peptide (referred to in FIG. 50 as T21) were also produced and tested, as shown in FIG. 50A-B .
  • FIG. 50A-B also presents data concerning blocked and unblocked peptides which contain additional amino acid residues from the gp41 region in which the DP107 sequence resides. Most of these peptides showed antiviral activity, as evidenced by their low IC 50 values.
  • EBV Zebra protein was synthesized utilizing SP6 RNA polymerase in vitro transcription and wheat germ in vitro translation systems (Promega Corporation recommendations; Butler, E. T. and Chamberlain, M. J., 1984, J. Biol. Chem. 257:5772; Pelham, H. R. B. and Jackson, R. J., 1976, Eur. J. Biochem. 67:247).
  • the in vitro translated Zebra protein was then preincubated with increasing amounts of peptide up to 250 ng/ml prior to the addition of 10,000 to 20,000 c.p.m. of a 32 P-labeled Zebra response element DNA fragment.
  • test peptide was assayed by the peptide's ability to abolish the response element gel migration retardation characteristic of a protein-bound nucleic acid molecule.
  • Peptides The peptides characterized in this study represent peptide walks through the region containing, and flanked on both sides by, the DP178/DP107 analog region identified in the Example presented in Section 20, above, and shown as shown in FIG. 33 . Specifically, the peptide walks covered the region from amino acid residue 173 to amino acid residue 246 of the EBV Zebra protein.
  • Each of the tested peptides were analyzed at a range of concentrations, with 150 ng/ml being the lowest concentration at which any of the peptides exerted an inhibitory effect.
  • the EBV Zebra protein transcription factor contains a DP178/DP107 analog region, as demonstrated in the Example presented, above, in Section 20. This protein appears to be the primary factor responsible for the reactivation capability of the virus. A method by which the DNA-binding function of the Zebra virus may be abolished may, therefore, represent an effective antiviral technique. In order to identify potential anti-EBV DP178/DP107 peptides, therefore, peptides derived from the region identified in Section 20, above, were tested for their ability to inhibit Zebra protein DNA binding.
  • the test peptides' ability to inhibit Zebra protein DNA binding was assayed via the EMSA assays described, above, in Section 28.1.
  • the data summarized in FIG. 51A-C presents the results of EMSA assays of the listed EBV test peptides. These peptides represent one amino acid “walks” through the region containing, and flanked on both sides by, the DP178/DP107 analog region identified in the Example presented in Section 20, above, and shown as shown in FIG. 33 .
  • the region from which these peptides are derived lies from EBV Zebra protein amino acid residue 173 to 246.
  • a number of the test peptides which were assayed exhibited an ability to inhibit Zebra protein homodimer DNA binding, including 439, 441, 444 and 445.
  • Those peptides which exhibit an ability to inhibit Zebra protein DNA binding represent potential anti-EBV antiviral compounds whose ability to inhibit EBV infection can be further characterized.

Abstract

The present invention relates to peptides which exhibit potent anti-retroviral activity. The peptides of the invention comprise DP178 (SEQ ID NO:1) peptide corresponding to amino acids 638 to 673 of the HIV-1LAI gp41 protein, and fragments, analogs and homologs of DP178. The invention further relates to the uses of such peptides as inhibitory of human and non-human retroviral, especially HIV, transmission to uninfected cells.

Description

    1. CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 08/484,223, filed Jun. 7, 1995, which is a division of U.S. patent application Ser. No. 08/470,896, filed Jun. 6, 1995, now U.S. Pat. No. 6,479,055, issued Nov. 12, 2002, which is a continuation-in-part of U.S. patent application Ser. No. 08/360,107, filed Dec. 20, 1994, now U.S. Pat. No. 6,017,536, issued Jan. 25, 2000, which is a continuation-in-part of U.S. patent application Ser. No. 08/255,208, filed Jun. 7, 1994, now U.S. Pat. No. 6,440,656, issued Aug. 27, 2002, each of which is incorporated by reference in its entirety. This application also incorporates by reference U.S. patent application Ser. No. 08/073,028, filed Jun. 7, 1993, now U.S. Pat. No. 5,464,933, issued Nov. 7, 1995.
  • This invention was made with Government support under Grant No. AI-30411-02 awarded by the National Institutes of Health. The Government has certain rights in the invention.
  • 2. INTRODUCTION
  • The present invention relates, first, to DP178 (SEQ ID NO:1), a peptide corresponding to amino acids 638 to 673 of the HIV-1LAI transmembrane protein (TM) gp41, and portions or analogs of DP178 (SEQ ID NO:1), which exhibit anti-membrane fusion capability, antiviral activity, such as the ability to inhibit HIV transmission to uninfected CD-4+ cells, or an ability to modulate intracellular processes involving coiled-coil peptide structures. Further, the invention relates to the use of DP178 (SEQ ID NO:1) and DP178 portions and/or analogs as antifusogenic or antiviral compounds or as inhibitors of intracellular events involving coiled-coil peptide structures. The present invention also relates to peptides analogous to DP107, a peptide corresponding to amino acids 558 to 595 of the HIV-1LAI transmembrane protein (TM) gp41, having amino acid sequences present in other viruses, such as enveloped viruses, and/or other organisms, and further relates to the uses of such peptides. These peptides exhibit anti-membrane fusion capability, antiviral activity, or the ability to modulate intracellular processes involving coiled-coil peptide structures. The present invention additionally relates to methods for identifying compounds that disrupt the interaction between DP178 and DP107, and/or between DP107-like and DP178-like peptides. Further, the invention relates to the use of the peptides of the invention as diagnostic agents. For example, a DP178 peptide may be used as an HIV subtype-specific diagnostic. The invention is demonstrated, first, by way of an Example wherein DP178 (SEQ ID NO:1), and a peptide whose sequence is homologous to DP178 are each shown to be potent, non-cytotoxic inhibitors of HIV-1 transfer to uninfected CD-4+ cells. The invention is further demonstrated by Examples wherein peptides having structural and/or amino acid motif similarity to DP107 and DP178 are identified in a variety of viral and nonviral organisms, and in examples wherein a number of such identified peptides derived from several different viral systems are demonstrated to exhibit antiviral activity.
  • 3. BACKGROUND OF THE INVENTION 3.1. Membrane Fusion Events
  • Membrane fusion is a ubiquitous cell biological process (for a review, see White, J. M., 1992, Science 258:917-924). Fusion events which mediate cellular housekeeping functions, such as endocytosis, constitutive secretion, and recycling of membrane components, occur continuously in all eukaryotic cells.
  • Additional fusion events occur in specialized cells. Intracellularly, for example, fusion events are involved in such processes as occur in regulated exocytosis of hormones, enzymes and neurotransmitters. Intercellularly, such fusion events feature prominently in, for example, sperm-egg fusion and myoblast fusion.
  • Fusion events are also associated with disease states. For example, fusion events are involved in the formation of giant cells during inflammatory reactions, the entry of all enveloped viruses into cells, and, in the case of human immunodeficiency virus (HIV), for example, are responsible for the virally induced cell-cell fusion which leads to cell death.
  • 3.2. The Human Immunodeficiency Virus
  • The human immunodeficiency virus (HIV) has been implicated as the primary cause of the slowly degenerative immune system disease termed acquired immune deficiency syndrome (AIDS) (Barre-Sinoussi, F. et al., 1983, Science 220:868-870; Gallo, R. et al., 1984, Science 224:500-503). There are at least two distinct types of HIV: HIV-1 (Barre-Sinoussi, F. et al., 1983, Science 220:868-870; Gallo R. et al., 1984, Science 224:500-503) and HIV-2 (Clavel, F. et al., 1986, Science 233:343-346; Guyader, M. et al., 1987, Nature 326:662-669). Further, a large amount of genetic heterogeneity exists within populations of each of these types. Infection of human CD-4+ T-lymphocytes with an HIV virus leads to depletion of the cell type and eventually to opportunistic infections, neurological dysfunctions, neoplastic growth, and ultimately death.
  • HIV is a member of the lentivirus family of retroviruses (Teich, N. et al., 1984, RNA Tumor Viruses, Weiss, R. et al., eds., CSH-Press, pp. 949-956). Retroviruses are small enveloped viruses that contain a diploid, single-stranded RNA genome, and replicate via a DNA intermediate produced by a virally-encoded reverse transcriptase, an RNA-dependent DNA polymerase (Varmus, H., 1988, Science 240:1427-1439). Other retroviruses include, for example, oncogenic viruses such as human T-cell leukemia viruses (HTLV-I, -II, -III), and feline leukemia virus.
  • The HIV viral particle consists of a viral core, composed of capsid proteins, that contains the viral RNA genome and those enzymes required for early replicative events. Myristylated Gag protein forms an outer viral shell around the viral core, which is, in turn, surrounded by a lipid membrane enveloped derived from the infected cell membrane. The HIV enveloped surface glycoproteins are synthesized as a single 160 Kd precursor protein which is cleaved by a cellular protease during viral budding into two glycoproteins, gp41 and gp120. gp41 is a transmembrane protein and gp120 is an extracellular protein which remains non-covalently associated with gp41, possibly in a trimeric or multimeric form (Hammarskjold, M. and Rekosh, D., 1989, Biochem. Biophys. Acta 989:269-280).
  • HIV is targeted to CD-4+ cells because the CD-4 cell surface protein acts as the cellular receptor for the HIV-1 virus (Dalgleish, A. et al., 1984, Nature 312:763-767; Klatzmann et al., 1984, Nature 312:767-768; Maddon et al., 1986, Cell 47:333-348). Viral entry into cells is dependent upon gp120 binding the cellular CD-4+ receptor molecules (McDougal, J. S. et al., 1986, Science 231:382-385; Maddon, P. J. et al., 1986, Cell 47:333-348) and thus explains HIV's tropism for CD-4+ cells, while gp41 anchors the enveloped glycoprotein complex in the viral membrane.
  • 3.3. HIV Treatment
  • HIV infection is pandemic and HIV associated diseases represent a major world health problem. Although considerable effort is being put into the successful design of effective therapeutics, currently no curative anti-retroviral drugs against AIDS exist. In attempts to develop such drugs, several stages of the HIV life cycle have been considered as targets for therapeutic intervention (Mitsuya, H. et al., 1991, FASEB J. 5:2369-2381). For example, virally encoded reverse transcriptase has been one focus of drug development. A number of reverse-transcriptase-targeted drugs, including 2′,3′-dideoxynucleoside analogs such as AZT, ddI, ddC, and d4T have been developed which have been shown to been active against HIV (Mitsuya. H. et al. 1991. Science 249:1533-1544). While beneficial, these nucleoside analogs are not curative, probably due to the rapid appearance of drug resistant HIV mutants (Lander, B. et al., 1989, Science 243:1731-1734). In addition, the drugs often exhibit toxic side effects such as bone marrow suppression, vomiting, and liver function abnormalities.
  • Attempts are also being made to develop drugs which can inhibit viral entry into the cell, the earliest stage of HIV infection. Here, the focus has thus far been on CD4, the cell surface receptor for HIV. Recombinant soluble CD4, for example, has been shown to inhibit infection of CD-4+ T-cells by some HIV-1 strains (Smith, D. H. et al., 1987, Science 238:1704-1707). Certain primary HIV-1 isolates, however, are relatively less sensitive to inhibition by recombinant CD-4 (Daar, E. et al., 1990, Proc. Natl. Acad. Sci. USA 87:6574-6579). In addition, recombinant soluble CD-4 clinical trials have produced inconclusive results (Schooley, R. et al., 1990, Ann. Int. Med. 112:247-253; Kahn, J. O. et al., 1990, Ann. Int. Med. 112:254-261; Yarchoan, R. et al., 1989, Proc. Vth Int. Conf. on AIDS, p. 564, MCP 137).
  • The late stages of HIV replication, which involve crucial virus-specific secondary processing of certain viral proteins, have also been suggested as possible anti-HIV drug targets. Late stage processing is dependent on the activity of a viral protease, and drugs are being developed which inhibit this protease (Erickson, J., 1990, Science 249:527-533). The clinical outcome of these candidate drugs is still in question.
  • Attention is also being given to the development of vaccines for the treatment of HIV infection. The HIV-1 enveloped proteins (gp160, gp120, gp41) have been shown to be the major antigens for anti-HIV antibodies present in AIDS patients (Barin, et al., 1985, Science 228:1094-1096). Thus far, therefore, these proteins seem to be the most promising candidates to act as antigens for anti-HIV vaccine development. To this end, several groups have begun to use various portions of gp160, gp120, and/or gp41 as immunogenic targets for the host immune system. See for example, Ivanoff, L. et al., U.S. Pat. No. 5,141,867; Saith, G. et al., WO 92/22,654; Shafferman, A., WO 91/09,872; Formoso, C. et al., WO 90/07,119. Clinical results concerning these candidate vaccines, however, still remain far in the future.
  • Thus, although a great deal of effort is being directed to the design and testing of anti-retroviral drugs, a truly effective, non-toxic treatment is still needed.
  • 4. SUMMARY OF THE INVENTION
  • The present invention relates, first, to DP178 (SEQ ID NO:1), a 36-amino acid synthetic peptide corresponding to amino acids 638 to 673 of the transmembrane protein (TM) gp41 from the HIV-1 isolate LAI (HIV-1LAI), which exhibits potent anti-HIV-1 activity. As evidenced by the Example presented below, in Section 6, the DP178 (SEQ ID NO:1) antiviral activity is so high that, on a weight basis, no other known anti-HIV agent is effective at concentrations as low as those at which DP178 (SEQ ID NO:1) exhibits its inhibitory effects.
  • The invention further relates to those portions and analogs of DP178 which also show such antiviral activity, and/or show anti-membrane fusion capability, or an ability to modulate intracellular processes involving coiled-coil peptide structures. The term “DP178 analog” refers to a peptide which contains an amino acid sequence corresponding to the DP178 peptide sequence present within the gp41 protein of HIV-1LAI, but found in viruses and/or organisms other than HIV-1LAI. Such DP178 analog peptides may, therefore, correspond to DP178-like amino acid sequences present in other viruses, such as, for example, enveloped viruses, such as retroviruses other than HIV-1LAI, as well as non-enveloped viruses. Further, such analogous DP178 peptides may also correspond to DP178-like amino acid sequences present in nonviral organisms.
  • The invention further relates to peptides DP107 analogs. DP107 is a peptide corresponding to amino acids 558-595 of the HIV-1LAI transmembrane protein (TM) gp41. The term “DP107 analog” as used herein refers to a peptide which contains an amino acid sequence corresponding to the DP107 peptide sequence present within the gp41 protein of HIV-1LAI, but found in viruses and organisms other than HIV-1LAI. Such DP107 analog peptides may, therefore, correspond to DP107-like amino acid sequences present in other viruses, such as, for example, enveloped viruses, such as retroviruses other than HIV-1LAI, as well as non-enveloped viruses. Further, such DP107 analog peptides may also correspond to DP107-like amino acid sequences present in nonviral organisms.
  • Further, the peptides of the invention include DP107 analog and DP178 analog peptides having amino acid sequences recognized or identified by the 107×178×4, ALLMOTI5 and/or PLZIP search motifs described herein.
  • The peptides of the invention may, for example, exhibit antifusogenic activity, antiviral activity, and/or may have the ability to modulate intracellular processes which involve coiled-coil peptide structures. With respect to the antiviral activity of the peptides of the invention, such an antiviral activity includes, but is not limited to the inhibition of HIV transmission to uninfected CD-4+ cells. Additionally, the antifusogenic capability, antiviral activity or intracellular modulatory activity of the peptides of the invention merely requires the presence of the peptides of the invention, and, specifically, does not require the stimulation of a host immune response directed against such peptides.
  • The peptides of the invention may be used, for example, as inhibitors of membrane fusion-asociated events, such as, for example, the inhibition of human and non-human retroviral, especially HIV, transmission to uninfected cells. It is further contemplated that the peptides of the invention may be used as modulators of intracellular events involving coiled-coil peptide structures.
  • The peptides of the invention may, alternatively, be used to identify compounds which may themselves exhibit antifusogenic, antiviral, or intracellular modulatory activity. Additional uses include, for example, the use of the peptides of the invention as organism or viral type and/or subtype-specific diagnostic tools.
  • The terms “antifusogenic” and “anti-membrane fusion”, as used herein, refer to an agent's ability to inhibit or reduce the level of membrane fusion events between two or more moieties relative to the level of membrane fusion which occurs between said moieties in the absence of the peptide. The moieties may be, for example, cell membranes or viral structures, such as viral envelopes or pili. The term “antiviral”, as used herein, refers to the compound's ability to inhibit viral infection of cells, via, for example, cell-cell fusion or free virus infection. Such infection may involve membrane fusion, as occurs in the case of enveloped viruses, or some other fusion event involving a viral structure and a cellular structure (e.g., such as the fusion of a viral pilus and bacterial membrane during bacterial conjugation). It is also contemplated that the peptides of the invention may exhibit the ability to modulate intracellular events involving coiled-coil peptide structures. “Modulate”, as used herein, refers to a stimulatory or inhibitory effect on the intracellular process of interest relative to the level or activity of such a process in the absence of a peptide of the invention.
  • Embodiments of the invention are demonstrated below wherein an extremely low concentration of DP178 (SEQ ID NO:1), and very low concentrations of a DP178 homolog (SEQ ID NO:3) are shown to be potent inhibitors of HIV-1 mediated CD-4+ cell-cell fusion (i.e., syncytial formation) and infection of CD-4+ cells by cell-free virus. Further, it is shown that DP178 (SEQ ID NO:1) is not toxic to cells, even at concentrations 3 logs higher than the inhibitory DP-178 (SEQ ID NO:1) concentration.
  • The present invention is based, in part, on the surprising discovery that the DP107 and DP178 domains of the HIV gp41 protein non-covalently complex with each other, and that their interaction is required for the normal infectivity of the virus. This discovery is described in the Example presented, below, in Section 8. The invention, therefore, further relates to methods for identifying antifusogenic, including antiviral, compounds that disrupt the interaction between DP107 and DP178, and/or between DP107-like and DP178-like peptides.
  • Additional embodiments of the invention (specifically, the Examples presents in Sections 9-16 and 19-25, below) are demonstrated, below, wherein peptides, from a variety of viral and nonviral sources, having structural and/or amino acid motif similarity to DP107 and DP178 are identified, and search motifs for their identification are described. Further, Examples (in Sections 17, 18, 25-29) are presented wherein a number of the peptides of the invention are demonstrated exhibit substantial antiviral activity or activity predictive of antiviral activity.
  • 4.1. DEFINITIONS
  • Peptides are defined herein as organic compounds comprising two or more amino acids covalently joined by peptide bonds. Peptides may be referred to with respect to the number of constituent amino acids, i.e., a dipeptide contains two amino acid residues, a tripeptide contains three, etc. Peptides containing ten or fewer amino acids may be referred to as oligopeptides, while those with more than ten amino acid residues are polypeptides. Such peptides may also include any of the modifications and additional amino and carboxy groups as are described herein.
  • Peptide sequences defined herein are represented by one-letter symbols for amino acid residues as follows:
      • A (alanine)
      • R (arginine)
      • N (asparagine)
      • D (aspartic acid)
      • C (cysteine)
      • Q (glutamine)
      • E (glutamic acid)
      • G (glycine)
      • H (histidine)
      • I (isoleucine)
      • L (leucine)
      • K (lysine)
      • M (methionine)
      • F (phenylalanine)
      • P (proline)
      • S (serine)
      • T (threonine)
      • W (tryptophan)
      • Y (tyrosine)
      • V (valine)
    5. BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. Amino acid sequence of DP178 (SEQ ID NO:1) derived from HIVLAI; DP178 homologs derived from HIV-1SF2 (DP-185; SEQ ID NO:3), HIV-1RF (SEQ ID NO:4), and HIV-1MN (SEQ ID NO:5); DP178 homologs derived from amino acid sequences of two prototypic HIV-2 isolates, namely, HIV-2rod (SEQ ID NO:6) and HIV-2NIHZ (SEQ ID NO:7); control peptides: DP-180 (SEQ ID NO:2), a peptide incorporating the amino acid residues of DP178 in a scrambled sequence; DP-118 (SEQ ID NO:10) unrelated to DP178, which inhibits HIV-1 cell free virus infection; DP-125 (SEQ ID NO:8), unrelated to DP178, also inhibits HIV-1 cell free virus infection; DP-116 (SEQ ID NO:9), unrelated to DP178, is negative for inhibition of HIV-1 infection when tested using a cell-free virus infection assay. Throughout the figures, the one letter amino acid code is used.
  • FIG. 2. Inhibition of HIV-1 cell-free virus infection by synthetic peptides. IC50 refers to the concentration of peptide that inhibits RT production from infected cells by 50% compared to the untreated control. Control: the level of RT produced by untreated cell cultures infected with the same level of virus as treated cultures.
  • FIG. 3. Inhibition of HIV-1 and HIV-2 cell-free virus infection by the synthetic peptide DP178 (SEQ ID NO:1). IC50: concentration of peptide that inhibits RT production by 50% compared to the untreated control. Control: Level of RT produced by untreated cell cultures infected with the same level of virus as treated cultures.
  • FIG. 4A-4B. Fusion Inhibition Assays. FIG. 4A: DP178 (SEQ ID NO:1) inhibition of HIV-1 prototypic isolate-mediated syncytial formation; data represents the number of virus-induced syncytial per cell. FIG. 4B: DP-180 (SEQ ID NO:2) represents a scrambled control peptide; DP-185 (SEQ ID NO:3) represents a DP178 homolog derived from HIV-1SF2 isolate; Control, refers to the number of syncytial produced in the absence of peptide.
  • FIG. 5. Fusion inhibition assay: HIV-1 vs. HIV-2. Data represents the number of virus-induced syncytial per well. ND: not done.
  • FIG. 6. Cytotoxicity study of DP178 (SEQ ID NO:1) and DP-116 (SEQ ID NO:9) on CEM cells. Cell proliferation data is shown.
  • FIG. 7. Schematic representation of HIV-gp41 and maltose binding protein (MBP)-gp41 fusion proteins. DP107 and DP178 are synthetic peptides based on the two putative helices of gp41. The letter P in the DP107 boxes denotes an Ile to Pro mutation at amino acid number 578. Amino acid residues are numbered according to Meyers et al., “Human Retroviruses and AIDS”, 1991, Theoret. Biol. and Biophys. Group, Los Alamos Natl. Lab., Los Alamos, N. Mex. The proteins are more fully described, below, in Section 8.1.1.
  • FIG. 8. A point mutation alters the conformation and anti-HIV activity of M41.
  • FIG. 9. Abrogation of DP178 anti-HIV activity. Cell fusion assays were carried out in the presence of 10 nM DP178 and various concentrations of M41Δ178 or M41PΔ178.
  • FIG. 10. Binding of DP178 to leucine zipper of gp41 analyzed by FAb-D ELISA.
  • FIG. 11A-B. Models for a structural transition in the HIV-1 TM protein. Two models are proposed which indicate a structural transition from a native oligomer to a fusogenic state following a trigger event (possibly gp120 binding to CD4). Common features of both models include (1) the native state is held together by noncovalent protein-protein interactions to form the heterodimer of gp120/41 and other interactions, principally though gp41 interactive sites, to form homo-oligomers on the virus surface of the gp120/41 complexes; (2) shielding of the hydrophobic fusogenic peptide at the N-terminus (F) in the native state; and (3) the leucine zipper domain (DP107) exists as a homo-oligomer coiled coil only in the fusogenic state. The major differences in the two models include the structural state (native or fusogenic) in which the DP107 and DP178 domains are complexed to each other. In the first model (FIG. 11A) this interaction occurs in the native state and in the second (FIG. 11B), it occurs during the fusogenic state. When triggered, the fusion complex in the model depicted in (A) is generated through formation of coiled-coil interactions in homologous DP107 domains resulting in an extended α-helix. This conformational change positions the fusion peptide for interaction with the cell membrane. In the second model (FIG. 11B), the fusogenic complex is stabilized by the association of the DP178 domain with the DP107 coiled-coil.
  • FIG. 12. Motif design using heptad repeat positioning of amino acids of known coiled-coils GCN4:(SEQ ID NO:84); C-FOS:(SEQ ID NO:85); C-JUN:(SEQ ID NO:86); C-MYC:(SEQ ID NO:87); FLU LOOP 36:(SEQ ID NO:88).
  • FIG. 13. Motif design using proposed heptad repeat positioning of amino acids of DP107 and DP178.
  • FIG. 14. Hybrid motif design crossing GCN4 and DP107.
  • FIG. 15. Hybrid motif design crossing GCN4 and DP178.
  • FIG. 16. Hybrid motif design 107×178×4, crossing DP107 (SEQ ID NO:89) and DP178 (SEQ ID NO:1). This motif was found to be the most consistent at identifying relevant DP107-like and DP178-like peptide regions.
  • FIG. 17. Hybrid motif design crossing GCN4, DP107, and DP178.
  • FIG. 18. Hybrid motif design ALLMOTI5 crossing GCN4, DP107, DP178, c-Fos c-Jun, c-Myc, and Flu Loop 36.
  • FIG. 19. PLZIP motifs designed to identify N-terminal proline-leucine zipper motifs.
  • FIG. 20. Search results for HIV-1 (BRU isolate) enveloped protein gp41 (SEQ ID NO:90). Sequence search motif designations: Spades (
    Figure US20100291680A1-20101118-P00001
    ): 107×178×4; Hearts (♡) ALLMOTI5; Clubs (
    Figure US20100291680A1-20101118-P00002
    ): PLZIP; Diamonds (♦): transmembrane region (the putative transmembrane domains were identified using a PC/Gene program designed to search for such peptide regions). Asterisk (*): Lupas method. The amino acid sequences identified by each motif are bracketed by the respective characters. Representative sequences chosen based on 107×178×4 searches are underlined and in bold. DP107 and DP178 sequences are marked, and additionally double-underlined and italicized.
  • FIG. 21. Search results for human respiratory syncytial virus (RSV) strain A2 fusion glycoprotein F1 (SEQ ID NO:91). Sequence search motif designations are as in FIG. 20.
  • FIG. 22. Search results for simian immunodeficiency virus (SIV) enveloped protein gp41 (AGM3 isolate) (SEQ ID NO:92). Sequence search motif designations are as in FIG. 20.
  • FIG. 23. Search results for canine distemper virus (strain Onderstepoort) fusion glycoprotein 1 (SEQ ID NO:93). Sequence search motif designations are as in FIG. 20.
  • FIG. 24. Search results for newcastle disease virus (strain Australia-Victoria/32) fusion glycoprotein F1 (SEQ ID NO:94). Sequence search motif designations are as in FIG. 20.
  • FIG. 25. Search results for human parainfluenza 3 virus (strain NIH 47885) fusion glycoprotein F1 (SEQ ID NO:95). Sequence search motif designations are as in FIG. 20.
  • FIG. 26. Search results for influenza A virus (strain A/AICHI/2/68) hemagglutinin precursor HA2 (SEQ ID NO:96). Sequence search designations are as in FIG. 20.
  • FIG. 27A-F. Respiratory Syncytial Virus (RSV) (SEQ ID NO:97) peptide antiviral and circular dichroism data. FIG. 27A-C: Peptides derived from the F2 DP178/DP107-like region T-22:(SEQ ID NO:121); T-68:(SEQ ID NO:122); T-334:(SEQ ID NO:123); T-371:(SEQ ID NO:124); T-372:(SEQ ID NO:125); T-373:(SEQ ID NO:126); T-374:(SEQ ID NO:127); T-375:(SEQ ID NO:128); T-575:(SEQ ID NO:129). Antiviral and CD data. FIG. 27D-F: Peptides derived from the F1 DP107-like region F1-107:(SEQ ID NO:98); T-12:(SEQ ID NO:130); T-13:(SEQ ID NO:131); T-15:(SEQ ID NO:132); T-19:(SEQ ID NO:133); T-28:(SEQ ID NO:134); T-30:(SEQ ID NO:135); T-66:(SEQ ID NO:136); T-576:(SEQ ID NO:137). Peptide and CD data.
  • Antiviral activity (AV) is represented by the following qualitative symbols:
      • “−”, negative antiviral activity;
      • “+/−”, antiviral activity at greater than 100 μg/ml;
      • “+”, antiviral activity at between 50-100 μg/ml;
      • “++”, antiviral activity at between 20-50 μg/ml;
      • “+++”, antiviral activity at between 1-20 μg/ml;
      • “++++”, antiviral activity at <1 μg/ml.
  • CD data, referring to the level of helicity is represented by the following qualitative symbol:
      • “−”, no helicity;
      • “+”, 25-50% helicity;
      • “++”, 50-75% helicity;
      • “+++” 75-100% helicity.
  • IC50 refers to the concentration of peptide necessary to produce only 50% of the number of syncytial relative to infected control cultures containing no peptide. IC50 values were obtained using purified peptides only.
  • FIG. 28A-C. Respiratory Syncytial Virus (RSV) DP178-like region (F1) peptide antiviral and CD data. Antiviral symbols, CD symbols, and IC50 are as in FIG. 27A-F. IC50 values were obtained using purified peptides only.
  • FIG. 29A-E. Peptides derived from the HPIV3 F1 DP107-like region. Peptide antiviral and CD data. Antiviral symbols, CD symbols, and IC50 are as in FIG. 27A-F. Purified peptides were used to obtain IC50 values, except where the values are marked by an asterisk (*), in which cases, the IC50 values were obtained using a crude peptide preparation.
  • FIG. 30A-C. Peptides derived from the HPIV3 F1 DP178-like region. Peptide antiviral and CD data. Antiviral symbols, CD symbols, and IC50 are as in FIG. 27A-F. Purified peptides were used to obtain IC50 values, except where the values are marked by an asterisk (*), in which cases, the IC50 values were obtained using a crude peptide preparation.
  • FIG. 31. Motif search results for simian immunodeficiency virus (SIV) isolate MM251, enveloped polyprotein gp41 (SEQ ID NO:102). Sequence search designations are as in FIG. 20.
  • FIG. 32. Motif search results for Epstein-Barr Virus (Strain B95-8), glycoprotein gp110 precursor (designated gp115) BALF4 (SEQ ID NO:103). Sequence search designations are as in FIG. 20.
  • FIG. 33. Motif search results for Epstein-Barr Virus (Strain B95-8), BZLF1 trans-activator protein (designated EB1 or Zebra) (SEQ ID NO:104). Sequence search designations are as in FIG. 20. Additionally, “@” refers to a well known DNA binding domain and “+” refers to a well known dimerization domain, as defined by Flemington and Speck (Flemington, E. and Speck, S. H., 1990, Proc. Natl. Acad. Sci. USA 87:9459-9463).
  • FIG. 34. Motif search results for measles virus (strain Edmonston), fusion glycoprotein F1 (SEQ ID NO:105). Sequence search designations are as in FIG. 20.
  • FIG. 35. Motif search results for Hepatitis B Virus (Subtype AYW), major surface antigen precursor S (SEQ ID NO:106). Sequence search designations are as in FIG. 20.
  • FIG. 36. Motif search results for simian Mason-Pfizer monkey virus, enveloped (TM) protein gp20 (SEQ ID NO:107). Sequence search designations are as in FIG. 20.
  • FIG. 37. Motif search results for Pseudomonas aerginosa, fimbrial protein (Pilin) (SEQ ID NO:108). Sequence search designations are as in FIG. 20.
  • FIG. 38. Motif search results for Neisseria gonorrhoeae fimbrial protein (Pilin) (SEQ ID NO:109). Sequence search designations are as in FIG. 20.
  • FIG. 39. Motif search results for Hemophilus influenzae fimbrial protein (SEQ ID NO:110). Sequence search designations are as in FIG. 20.
  • FIG. 40. Motif search results for Staphylococcus aureus, toxic shock syndrome toxin-1 (SEQ ID NO:111). Sequence search designations are as in FIG. 20.
  • FIG. 41. Motif search results for Staphylococcus aureus enterotoxin Type E (SEQ ID NO:112). Sequence search designations are as in FIG. 20.
  • FIG. 42. Motif search results for Staphylococcus aureus enterotoxin A (SEQ ID NO:113). Sequence search designations are as in FIG. 20.
  • FIG. 43. Motif search results for Escherichia coli, heat labile enterotoxin A (SEQ ID NO:114). Sequence search designations are as in FIG. 20.
  • FIG. 44. Motif search results for human c-fos proto-oncoprotein (SEQ ID NO:115). Sequence search designations are as in FIG. 20.
  • FIG. 45. Motif search results for human lupus KU autoantigen protein P70 (SEQ ID NO:116). Sequence search designations are as in FIG. 20.
  • FIG. 46. Motif search results for human zinc finger protein 10 (SEQ ID NO:117). Sequence search designations are as in FIG. 20.
  • FIG. 47A-B. Measles virus (MeV) fusion protein DP178-like region antiviral and CD data T-252AO:(SEQ ID NO:118); T-268AO:(SEQ ID NO:119). Antiviral symbols, CD symbols, and IC50 are as in FIG. 27A-F. IC50 values were obtained using purified peptides.
  • FIG. 48A-B. Simian immunodeficiency virus (SIV) TM (fusion) protein DP178-like region antiviral data (SEQ ID NO:120). Antiviral symbols are as in FIG. 27A-F “NT”, not tested.
  • FIG. 49A-L. DP178-derived peptide antiviral data [(SEQ ID NO:158); T50:(SEQ ID NO:159); (SEQ ID NO:160); T234:(SEQ ID NO:161); T235:(SEQ ID NO:162); T570:(SEQ ID NO:163); T381:(SEQ ID NO:164); T677:(SEQ ID NO:165); T589:(SEQ ID NO:166); T590:(SEQ ID NO:167); T591:(SEQ ID NO:168); T270:(SEQ ID NO:169); T271:(SEQ ID NO:170); T273:(SEQ ID NO:171); T608:(SEQ ID NO:172); T609:(SEQ ID NO:173); T610:(SEQ ID NO:174); T611:(SEQ ID NO:175); T612:(SEQ ID NO:176); T595:(SEQ ID NO:177); T95:(SEQ ID NO:178); T96:(SEQ ID NO:179); T97:(SEQ ID NO:180); T98:(SEQ ID NO:181); T99:(SEQ ID NO:182); T103:(SEQ ID NO:183); T212:(SEQ ID NO:184); T213:(SEQ ID NO:185); T214:(SEQ ID NO:186); T215:(SEQ ID NO:187); T216:(SEQ ID NO:188); T229:(SEQ ID NO:189); T230:(SEQ ID NO:190); T231:(SEQ ID NO:191); T379:(SEQ ID NO:192); T701:(SEQ ID NO:193); T702:(SEQ ID NO:194); T703:(SEQ ID NO:195); T704:(SEQ ID NO:196); T705:(SEQ ID NO:197); T706:(SEQ ID NO:198); T156:(SEQ ID NO:199); T90:(SEQ ID NO:200)]. The peptides listed herein were derived from the region surrounding the HIV-1 BRU isolate DP178 region (e.g., gp41 amino acid residues 615-717).
  • In instances where peptides contained DP178 point mutations, the mutated amino acid residues are shown with a shaded background. In instances in which the test peptide has had an amino and/or carboxy-terminal group added or removed (apart from the standard amido- and acetyl-blocking groups found on such peptides), such modifications are indicated. FIG. 49A-D: The column to the immediate right of the name of the test peptide indicates the size of the test peptide and points out whether the peptide is derived from a one amino acid peptide “walk” across the DP178 region. The next column to the right indicates whether the test peptide contains a point mutation, while the column to its right indicates whether certain amino acid residues have been added to or removed from the DP178-derived amino acid sequence. FIG. 49E-H: The column to the immediate right of the test peptide name indicates whether the peptide represents a DP178 truncation, the next column to the right points out whether the peptide contains a point mutation, and the column to its right indicates whether the peptide contains amino acids which have been added to or removed from the DP178 sequence itself FIG. 49I-L: The column to the immediate right of the test peptide name indicates whether the test peptide contains a point mutation, while the column to its right indicates whether amino acid residues have been added to or removed from the DP178 sequence itself. IC50 is as defined in FIG. 27A-F, and IC50 values were obtained using purified peptides except where marked with an asterisk (*), in which case the IC50 was obtained using a crude peptide preparation.
  • FIG. 50A-B. DP107 and DP107 gp41 region truncated peptide antiviral data (SEQ ID NO:201). IC50 as defined in FIG. 27A-D, and IC50 values were obtained using purified peptides except where marked with an asterisk (*), in which case the IC50 was obtained using a crude peptide preparation.
  • FIG. 51A-C. Epstein-Barr virus Strain B95-8 BZLF1 DP178/DP107 analog region peptide walks and electrophoretic mobility shift assay results. The peptides 173-219:(SEQ ID NO:202); 185-230:(SEQ ID NO:203); T-446:(SEQ ID NO:204); 197-242:(SEQ ID NO:205); T-458:(SEQ ID NO:206); 209-246:(SEQ ID NO:207) (T-423 to T-446, FIG. 51A-B; T-447 to T-461, FIG. 51C) represent one amino acid residue “walks” through the EBV Zebra protein region from amino acid residue 173 to 246.
  • The amino acid residue within this region which corresponds to the first amino acid residue of each peptide is listed to the left of each peptide, while the amino acid residue within this region which corresponds to the last amino acid residue of each peptide is listed to the right of each peptide. The length of each test peptide is listed at the far right of each line, under the heading “Res”.
  • “ACT” refers to a test peptide's ability to inhibit Zebra binding to its response element. “+” refers to a visible, but incomplete, abrogation of the response element/Zebra homodimer complex; “+++” refers to a complete abrogation of the complex; and “−” represents a lack of complex disruption.
  • FIG. 52A-B. Hepatitis B virus subtype AYW major surface antigen precursor S protein DP178/DP107 analog region and peptide walks. 52A depicts Domain I (SEQ ID NO:208) (S protein amino acid residues 174-219), which contains a potential DP178/DP107 analog region. In addition, peptides are listed which represent one amino acid peptide “walks” through domain I. 52B depicts Domain II (SEQ ID NO:209) (S protein amino acid residues 233-290), which contains a second potential DP178/DP107 analog region. In addition, peptides are listed which represent one amino acid peptide “walks” through domain II.
  • 6. DETAILED DESCRIPTION OF THE INVENTION
  • Described herein are peptides which may exhibit antifusogenic activity, antiviral capability, and/or the ability to modulate intracellular processes involving coiled-coil peptide structures. The peptides described include, first, DP178 (SEQ ID NO:1), a gp41-derived 36 amino acid peptide and fragments and analogs of DP178.
  • In addition, the peptides of the invention described herein include peptides which are DP107 analogs. DP107 (SEQ ID NO:99) is a 38 amino acid peptide corresponding to residues 558 to 595 of the HIV-1LAI transmembrane (TM) gp41 protein. Such DP107 analogs may exhibit antifusogenic capability, antiviral activity or an ability to modulate intracellular processes involving coiled-coil structures.
  • Further, peptides of the invention include DP107 and DP178 are described herein having amino acid sequences recognized by the 107×178×4, ALLMOTI5, and PLZIP search motifs. Such motifs are also discussed.
  • Also described here are antifusogenic, antiviral, intracellular modulatory, and diagnostic uses of the peptides of the invention. Further, procedures are described for the use of the peptides of the invention for the identification of compounds exhibiting antifusogenic, antiviral or intracellular modulatory activity.
  • While not limited to any theory of operation, the following model is proposed to explain the potent anti-HIV activity of DP178, based, in part, on the experiments described in the Examples, infra. In the HIV protein, gp41, DP178 corresponds to a putative α-helix region located in the C-terminal end of the gp41 ectodomain, and appears to associate with a distal site on gp41 whose interactive structure is influenced by the leucine zipper motif, a coiled-coil structure, referred to as DP107. The association of these two domains may reflect a molecular linkage or “molecular clasp” intimately involved in the fusion process. It is of interest that mutations in the C-terminal α-helix motif of gp41 (i.e., the D178 domain) tend to enhance the fusion ability of gp41, whereas mutations in the leucine zipper region (i.e., the DP107 domain) decrease or abolish the fusion ability of the viral protein. It may be that the leucine zipper motif is involved in membrane fusion while the C-terminal α-helix motif serves as a molecular safety to regulate the availability of the leucine zipper during virus-induced membrane fusion.
  • On the basis of the foregoing, two models are proposed of gp41-mediated membrane fusion which are schematically shown in FIG. 11A-B. The reason for proposing two models is that the temporal nature of the interaction between the regions defined by DP107 and DP178 cannot, as yet, be pinpointed. Each model envisions two conformations for gp41-one in a “native” state as it might be found on a resting virion. The other in a “fusogenic” state to reflect conformational changes triggered following binding of gp120 to CD4 and just prior to fusion with the target cell membrane. The strong binding affinity between gp120 and CD4 may actually represent the trigger for the fusion process obviating the need for a pH change such as occurs for viruses that fuse within intracellular vesicles. The two major features of both models are: (1) the leucine zipper sequences (DP107) in each chain of oligomeric enveloped are held apart in the native state and are only allowed access to one another in the fusogenic state so as to form the extremely stable coiled-coils, and (2) association of the DP178 and DP107 sites as they exist in gp41 occur either in the native or fusogenic state. FIG. 11A depicts DP178/DP107 interaction in the native state as a molecular clasp. On the other hand, if one assumes that the most stable form of the enveloped occurs in the fusogenic state, the model in FIG. 11B can be considered.
  • When synthesized as peptides, both DP107 and DP178 are potent inhibitors of HIV infection and fusion, probably by virtue of their ability to form complexes with viral gp41 and interfere with its fusogenic process; e.g., during the structural transition of the viral protein from the native structure to the fusogenic state, the DP178 and DP107 peptides may gain access to their respective binding sites on the viral gp41, and exert a disruptive influence. DP107 peptides which demonstrate anti-HIV activity are described in Applicants' co-pending application Ser. No. 08/264,531, filed Jun. 23, 1994, which is incorporated by reference herein in its entirety.
  • As shown in the Examples, infra, a truncated recombinant gp41 protein corresponding to the ectodomain of gp41 containing both DP107 and DP178 domains (excluding the fusion peptide, transmembrane region and cytoplasmic domain of gp41) did not inhibit HIV-1 induced fusion. However, when a single mutation was introduced to disrupt the coiled-coil structure of the DP107 domain—a mutation which results in a total loss of biological activity of DP107 peptides—the inactive recombinant protein was transformed to an active inhibitor of HIV-1 induced fusion. This transformation may result from liberation of the potent DP178 domain from a molecular clasp with the leucine zipper, DP107 domain.
  • For clarity of discussion, the invention will be described primarily for DP178 peptide inhibitors of HIV. However, the principles may be analogously applied to other viruses, both enveloped and nonenveloped, and to other non-viral organisms.
  • 6.1. DP178 and DP178-Like Peptides
  • The DP178 peptide (SEQ ID NO:1) of the invention corresponds to amino acid residues 638 to 673 of the transmembrane protein gp41 from the HIV-1LAI isolate, and has the 36 amino acid sequence (reading from amino to carboxy terminus):
  • (SEQ ID NO: 1)
    NH2-YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF-COOH
  • In addition to the full-length DP178 (SEQ ID NO:1) 36-mer, the peptides of the invention may include truncations of the DP178 (SEQ ID NO:1) peptide which exhibit antifusogenic activity, antiviral activity and/or the ability to modulate intracellular processes involving coiled-coil peptide structures. Truncations of DP178 (SEQ ID NO:1) peptides may comprise peptides of between 3 and 36 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 36-mer polypeptide), as shown in Tables I and IA, below. Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus. “X” may represent an amino group (—NH2) and “Z” may represent a carboxyl (—COOH) group. Alternatively, “X” may represent a hydrophobic group, including but not limited to carbobenzyl, dansyl, or T-butoxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group. Further, “Z” may represent an amido group; a T-butoxycarbonyl group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group. A preferred “X” or “Z” macromolecular group is a peptide group.
  • TABLE I
    DP178 (SEQ ID NO: 1) CARBOXY TRUNCATIONS
    X-YTS-Z
    X-YTSL-Z
    X-YTSLI-Z
    X-YTSLIH-Z
    X-YTSLIHS-Z
    X-YTSLIHSL-Z
    X-YTSLIHSLI-Z
    X-YTSLIHSLIE-Z
    X-YTSLIHSLIEE-Z
    X-YTSLIHSLIEES-Z
    X-YTSLIHSLIEESQ-Z
    X-YTSLIHSLIEESQN-Z
    X-YTSLIHSLIEESQNQ-Z
    X-YTSLIHSLIEESQNQQ-Z
    X-YTSLIHSLIEESQNQQE-Z
    X-YTSLIHSLIEESQNQQEK-Z
    X-YTSLIHSLIEESQNQQEKN-Z
    X-YTSLIHSLIEESQNQQEKNE-Z
    X-YTSLIHSLIEESQNQQEKNEQ-Z
    X-YTSLIHSLIEESQNQQEKNEQE-Z
    X-YTSLIHSLIEESQNQQEKNEQEL-Z
    X-YTSLIHSLIEESQNQQEKNEQELL-Z
    X-YTSLIHSLIEESQNQQEKNEQELLE-Z
    X-YTSLIHSLIEESQNQQEKNEQELLEL-Z
    X-YTSLIHSLIEESQNQQEKNEQELLELD-Z
    X-YTSLIHSLIEESQNQQEKNEQELLELDK-Z (SEQ ID NO: 260)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKW-Z (SEQ ID NO: 259)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWA-Z (SEQ ID NO: 258)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWAS-Z (SEQ ID NO: 257)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWASL-Z (SEQ ID NO: 256)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWASLW-Z (SEQ ID NO: 255)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWASLWN-Z (SEQ ID NO: 254)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNW-Z (SEQ ID NO: 253)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 1)
    The one letter amino acid code is used.
    Additionally,“X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
    “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • TABLE IA
    DP178 (SEQ ID NO: 1) AMINO TRUNCATIONS
    X-NWF-Z
    X-WNWF-Z
    X-LWNWF-Z
    X-SLWNWF-Z
    X-ASLWNWF-Z
    X-WASLWNWF-Z
    X-KWASLWNWF-Z
    X-DKWASLWNWF-Z
    X-LDKWASLWNWF-Z
    X-ELDKWASLWNWF-Z
    X-LELDKWASLWNWF-Z
    X-LLELDKWASLWNWF-Z
    X-ELLELDKWASLWNWF-Z
    X-QELLELDKWASLWNWF-Z
    X-EQELLELDKWASLWNWF-Z
    X-NEQELLELDKWASLWNWF-Z
    X-KNEQELLELDKWASLWNWF-Z
    X-EKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 238)
    X-QEKNEQELLELDKWASLWNWF-Z
    X-QQEKNEQELLELDKWASLWNWF-Z
    X-NQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 237)
    X-QNQQEKNEQELLELDKWASLWNWF-Z
    X-SQNQQEKNEQELLELDKWASLWNWF-Z
    X-ESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 236)
    X-EESQNQQEKNEQELLELDKWASLWNWF-Z
    X-IEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 268)
    X-LIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 267)
    X-SLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 266)
    X-HSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 265)
    X-IHSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 264)
    X-LIHSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 263)
    X-SLIHSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 262)
    X-TSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 261)
    X-YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF-Z (SEQ ID NO: 1)
    The one letter amino acid code is used.
    Additionally, “X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
    “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • The peptides of the invention also include DP178-like peptides. “DP178-like”, as used herein, refers, first, to DP178 and DP178 truncations which contain one or more amino acid substitutions, insertions and/or deletions. Second, “DP-178-like” refers to peptide sequences identified or recognized by the ALLMOTI5, 107×178×4 and PLZIP search motifs described herein, having structural and/or amino acid motif similarity to DP178. The DP178-like peptides of the invention may exhibit antifusogenic or antiviral activity, or may exhibit the ability to modulate intracellular processes involving coiled-coil peptides. Further, such DP178-like peptides may possess additional advantageous features, such as, for example, increased bioavailability, and/or stability, or reduced host immune recognition.
  • HIV-1 and HIV-2 enveloped proteins are structurally distinct, but there exists a striking amino acid conservation within the DP178-corresponding regions of HIV-1 and HIV-2. The amino acid conservation is of a periodic nature, suggesting some conservation of structure and/or function. Therefore, one possible class of amino acid substitutions would include those amino acid changes which are predicted to stabilize the structure of the DP178 peptides of the invention. Utilizing the DP178 and DP178 analog sequences described herein, the skilled artisan can readily compile DP178 consensus sequences and ascertain from these, conserved amino acid residues which would represent preferred amino acid substitutions.
  • The amino acid substitutions may be of a conserved or non-conserved nature. Conserved amino acid substitutions consist of replacing one or more amino acids of the DP178 (SEQ ID NO:1) peptide sequence with amino acids of similar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to aspartic acid (D) amino acid substitution. Non-conserved substitutions consist of replacing one or more amino acids of the DP178 (SEQ ID NO:1) peptide sequence with amino acids possessing dissimilar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to valine (V) substitution.
  • Amino acid insertions may consist of single amino acid residues or stretches of residues. The insertions may be made at the carboxy or amino terminal end of the DP178 or DP178 truncated peptides, as well as at a position internal to the peptide. Such insertions will generally range from 2 to 15 amino acids in length. It is contemplated that insertions made at either the carboxy or amino terminus of the peptide of interest may be of a broader size range, with about 2 to about 50 amino acids being preferred. One or more such insertions may be introduced into DP178 (SEQ. ID NO:1) or DP178 truncations, as long as such insertions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • Preferred amino or carboxy terminal insertions are peptides ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP178 gp41 amino acid sequence, respectively. Thus, a preferred amino terminal or carboxy terminal amino acid insertion would contain gp41 amino acid sequences found immediately amino to or carboxy to the DP178 region of the gp41 protein.
  • Deletions of DP178 (SEQ ID NO:1) or DP178 truncations are also within the scope of the invention. Such deletions consist of the removal of one or more amino acids from the DP178 or DP178-like peptide sequence, with the lower limit length of the resulting peptide sequence being 4 to 6 amino acids. Such deletions may involve a single contiguous or greater than one discrete portion of the peptide sequences. One or more such deletions may be introduced into DP178 (SEQ ID NO:1) or DP178 truncations, as long as such deletions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • DP178 analogs are further described, below, in Section 5.3.
  • 6.1.1 DP107 and DP107-Like Peptides
  • Further, the peptides of the invention include peptides having amino acid sequences corresponding to DP107 analogs. DP107 is a 38 amino acid peptide which exhibits potent antiviral activity, and corresponds to residues 558 to 595 of HIV-1LAI transmembrane (TM) gp41 protein, as shown here:
  • (SEQ ID NO: 89)
    NH2-NNLLRAIEAQQHLLQLTVWQIKQLQARILAVERYLKDQ-COOH
  • In addition to the full-length DP107 38-mer, the peptides of the invention may include truncations of the DP107 peptide which exhibit antifusogenic activity, antiviral activity and/or the ability to modulate intracellular processes involving coiled-coil peptide structures. Truncations of DP107 peptides may comprise peptides of between 3 and 38 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 38-mer polypeptide), as shown in Tables II and IIA, below. Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus. “X” may represent an amino group (—NH2) and “Z” may represent a carboxyl (—COOH) group. Alternatively, “X” may represent a hydrophobic group, including but not limited to carbobenzyl, dansyl, or T-butoxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group. Further, “Z” may represent an amido group; a T-butoxycarbonyl group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group. A preferred “X” or “Z” macromolecular group is a peptide group.
  • TABLE II
    DP107 (SEQ ID NO: 89) CARBOXY TRUNCATIONS X-NNL-Z
    X-NNLL-Z
    X-NNLLR-Z
    X-NNLLRA-Z
    X-NNLLRAI-Z
    X-NNLLRAIE-Z
    X-NNLLRAIEA-Z
    X-NNLLRAIEAQ-Z
    X-NNLLRAIEAQQ-Z
    X-NNLLRAIEAQQH-Z
    X-NNLLRAIEAQQHL-Z
    X-NNLLRAIEAQQHLL-Z
    X-NNLLRAIEAQQHLLQ-Z
    X-NNLLRAIEAQQHLLQL-Z
    X-NNLLRAIEAQQHLLQLT-Z
    X-NNLLRAIEAQQHLLQLTV-Z
    X-NNLLRAIEAQQHLLQLTVW-Z
    X-NNLLRAIEAQQHLLQLTVWG-Z
    X-NNLLRAIEAQQHLLQLTVWGI-Z
    X-NNLLRAIEAQQHLLQLTVWGIK-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQ-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQL-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQ-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQA-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQAR-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARI-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARIL-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILA-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAV-Z (SEQ ID NO: 243)
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVE-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVER-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVERY-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVERYL-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVERYLK-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVERYLKD-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z (SEQ ID NO: 89)
    The one letter amino acid code is used.
    Additionally, “X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
    “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • TABLE IIA
    DP107 (SEQ ID NO: 89) AMINO TRUNCATIONS
    X-KDQ-Z (SEQ ID NO: 89)
    X-LKDQ-Z
    X-YLKDQ-Z
    X-RYLKDQ-Z
    X-ERYLKDQ-Z
    X-VERYLKDQ-Z
    X-AVERYLKDQ-Z
    X-LAVERYLKDQ-Z
    X-ILAVERYLKDQ-Z
    X-RILAVERYLKDQ-Z
    X-ARILAVERYLKDQ-Z
    X-QARILAVERYLKDQ-Z
    X-LQARILAVERYLKDQ-Z
    X-QLQARILAVERYLKDQ-Z
    X-KQLQARILAVERYLKDQ-Z
    X-IKQLQARILAVERYLKDQ-Z
    X-GIKQLQARILAVERYLKDQ-Z
    X-WGIKQLQARILAVERYLKDQ-Z
    X-VWGIKQLQARILAVERYLKDQ-Z
    X-TVWGIKQLQARILAVERYLKDQ-Z
    X-LTVWGIKQLQARILAVERYLKDQ-Z
    X-QLTVWGIKQLQARILAVERYLKDQ-Z
    X-LQLTVWGIKQLQARILAVERYLKDQ-Z
    X-LLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-HLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-QHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-QQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-AQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-EAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-IEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-AIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-RAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-LRAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-LLRAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-NLLRAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    X-NNLLRAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ-Z
    The one letter amino acid code is used.
    Additionally, “X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
    “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • The peptides of the invention also include DP107-like peptides. “DP107-like”, as used herein, refers, first, to DP107 and DP107 truncations which contain one or more amino acid substitutions, insertions and/or deletions. Second, “DP-107-like” refers to peptide sequences identified or recognized by the ALLMOTI5, 107×178×4 and PLZIP search motifs described herein, having structural and/or amino acid motif similarity to DP107. The DP107-like peptides of the invention may exhibit antifusogenic or antiviral activity, or may exhibit the ability to modulate intracellular processes involving coiled-coil peptides. Further, such DP107-like peptides may possess additional advantageous features, such as, for example, increased bioavailability, and/or stability, or reduced host immune recognition.
  • HIV-1 and HIV-2 enveloped proteins are structurally distinct, but there exists a striking amino acid conservation within the DP107-corresponding regions of HIV-1 and HIV-2. The amino acid conservation is of a periodic nature, suggesting some conservation of structure and/or function. Therefore, one possible class of amino acid substitutions would include those amino acid changes which are predicted to stabilize the structure of the DP107 peptides of the invention. Utilizing the DP107 and DP107 analog sequences described herein, the skilled artisan can readily compile DP107 consensus sequences and ascertain from these, conserved amino acid residues which would represent preferred amino acid substitutions.
  • The amino acid substitutions may be of a conserved or non-conserved nature. Conserved amino acid substitutions consist of replacing one or more amino acids of the DP107 peptide sequence with amino acids of similar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to aspartic acid (D) amino acid substitution. Non-conserved substitutions consist of replacing one or more amino acids of the DP107 peptide sequence with amino acids possessing dissimilar charge, size, and/or hydrophobicity characteristics, such as, for example, a glutamic acid (E) to valine (V) substitution.
  • Amino acid insertions may consist of single amino acid residues or stretches of residues. The insertions may be made at the carboxy or amino terminal end of the DP107 or DP107 truncated peptides, as well as at a position internal to the peptide. Such insertions will generally range from 2 to 15 amino acids in length. It is contemplated that insertions made at either the carboxy or amino terminus of the peptide of interest may be of a broader size range, with about 2 to about 50 amino acids being preferred. One or more such insertions may be introduced into DP107 or DP107 truncations, as long as such insertions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • Preferred amino or carboxy terminal insertions are peptides ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP107 gp41 amino acid sequence, respectively. Thus, a preferred amino terminal or carboxy terminal amino acid insertion would contain gp41 amino acid sequences found immediately amino to or carboxy to the DP107 region of the gp41 protein.
  • Deletions of DP107 or DP178 truncations are also within the scope of the invention. Such deletions consist of the removal of one or more amino acids from the DP107 or DP107-like peptide sequence, with the lower limit length of the resulting peptide sequence being 4 to 6 amino acids. Such deletions may involve a single contiguous or greater than one discrete portion of the peptide sequences. One or more such deletions may be introduced into DP107 or DP107 truncations, as long as such deletions result in peptides which may still be recognized by the 107×178×4, ALLMOTI5 or PLZIP search motifs described herein, or may, alternatively, exhibit antifusogenic or antiviral activity, or exhibit the ability to modulate intracellular processes involving coiled-coil peptide structures.
  • DP107 and DP107 truncations are more fully described in Applicants' co-pending U.S. patent application Ser. No. 08/374,666, filed Jan. 27, 1995, and which is incorporated herein by reference in its entirety. DP107 analogs are further described, below, in Section 5.3.
  • 6.1.2 DP107 and DP178 Analogs
  • Peptides corresponding to analogs of the DP178, DP178 truncations, DP107 and DP107 truncation sequences of the invention, described, above, in Sections 5.1 and 5.2 may be found in other viruses, including, for example, non-HIV-1LAI enveloped viruses, non-enveloped viruses and other non-viral organisms.
  • The term “analog”, as used herein, refers to a peptide which is recognized or identified via the 107×178×4, ALLMOTI5 and/or PLZIP search strategies discussed below. Further, such peptides may exhibit antifusogenic capability, antiviral activity, or the ability to modulate intracellular processes involving coiled-coil structures.
  • Such DP178 and DP107 analogs may, for example, correspond to peptide sequences present in TM proteins of enveloped viruses and may, additionally correspond to peptide sequences present in non enveloped and non-viral organisms. Such peptides may exhibit anti fusogenic activity, antiviral activity, most particularly antiviral activity which is specific to the virus in which their native sequences are found, or may exhibit an ability to modulate intracellular processes involving coiled-coil peptide structures.
  • DP178 analogs are peptides whose amino acid sequences are comprised of the amino acid sequences of peptide regions of, for example, other (i.e., other than HIV-1LAI) viruses that correspond to the gp41 peptide region from which DP178 (SEQ ID:1) was derived. Such viruses may include, but are not limited to, other HIV-1 isolates and HIV-2 isolates. DP178 analogs derived from the corresponding gp41 peptide region of other (i.e., non HIV-1LAI) HIV-1 isolates may include, for example, peptide sequences as shown below.
  • (DP-185; SEQ ID NO: 3)
    NH2-YTNTIYTLLEESQNQQEKNEQELLELDKWASLWNWF-COOH;
    (SEQ ID NO: 4)
    NH2-YTGIIYNLLEESQNQQEKNEQELLELDKWANLWNWF-COOH;
    (SEQ ID NO: 5)
    NH2-YTSLIYSLLEKSQIQQEKNEQELLELDKWASLWNWF-COOH.

    SEQ ID NO:3 (DP-185), SEQ ID NO:4, and SEQ ID NO:5 are derived from HIV-1SF2, HIV-1RF, and HIV-1MN isolates, respectively. Underlined amino acid residues refer to those residues that differ from the corresponding position in the DP178 (SEQ ID NO:1) peptide. One such DP178 analog, DP-185 (SEQ ID NO:3), is described in the Example presented in Section 6, below, where it is demonstrated that DP-185 (SEQ ID NO:3) exhibits antiviral activity. The DP178 analogs of the invention may also include truncations, as described above. Further, the analogs of the invention modifications such those described for DP178 analogs in Section 5.1., above. It is preferred that the DP178 analogs of the invention represent peptides whose amino acid sequences correspond to the DP178 region of the gp41 protein, it is also contemplated that the peptides of the invention may, additionally, include amino sequences, ranging from about 2 to about 50 amino acid residues in length, corresponding to gp41 protein regions either amino to or carboxy to the actual DP178 amino acid sequence.
  • Striking similarities, as shown in FIG. 1, exist within the regions of HIV-1 and HIV-2 isolates which correspond to the DP178 sequence. A DP178 analog derived from the HIV-2NIHZ isolate has the 36 amino acid sequence (reading from amino to carboxy terminus):
  • (SEQ ID NO: 7)
    NH2-LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-COOH
  • Table III and Table IV show some possible truncations of the HIV-2NIHZ DP178 analog, which may comprise peptides of between 3 and 36 amino acid residues (i.e., peptides ranging in size from a tripeptide to a 36-mer polypeptide). Peptide sequences in these tables are listed from amino (left) to carboxy (right) terminus. “X” may represent an amino group (—NH2) and “Z” may represent a carboxyl (—COOH) group. Alternatively, “X” may represent a hydrophobic group, including but not limited to carbobenzyl, dansyl, or T-butoxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group. Further, “Z” may represent an amido group; a T-butoxycarbonyl group; or a covalently attached macromolecular group, including but not limited to a lipid-fatty acid conjugate, polyethylene glycol, carbohydrate or peptide group. A preferred “X” or “Z” macromolecular group is a peptide group.
  • TABLE III
    HIV-2NIHZ DP178 ANALOG CARBOXY TRUNCATIONS
    X-LEA-Z (SEQ ID NO: 7)
    X-LEAN-Z
    X-LEANI-Z
    X-LEANIS-Z
    X-LEANISQ-Z
    X-LEANISQS-Z
    X-LEANISQSL-Z
    X-LEANISQSLE-Z
    X-LEANISQSLEQ-Z
    X-LEANISQSLEQA-Z
    X-LEANISQSLEQAQ-Z
    X-LEANISQSLEQAQI-Z
    X-LEANISQSLEQAQIQ-Z
    X-LEANISQSLEQAQIQQ-Z
    X-LEANISQSLEQAQIQQE-Z
    X-LEANISQSLEQAQIQQEK-Z
    X-LEANISQSLEQAQIQQEKN-Z
    X-LEANISQSLEQAQIQQEKNM-Z
    X-LEANISQSLEQAQIQQEKNMY-Z
    X-LEANISQSLEQAQIQQEKNMYE-Z
    X-LEANISQSLEQAQIQQEKNMYEL-Z
    X-LEANISQSLEQAQIQQEKNMYELQ-Z
    X-LEANISQSLEQAQIQQEKNMYELQK-Z
    X-LEANISQSLEQAQIQQEKNMYELQKL-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLN-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNS-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSW-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWD-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDV-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDVF-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDVFT-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTN-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNW-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    The one letter amino acid code is used.
    Additionally, “X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
    “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • TABLE IV
    HIV-2NIHZ DP178 ANALOG AMINO TRUNCATIONS
    X-NWL-Z (SEQ ID NO: 7)
    X-TNWL-Z
    X-FTNWL-Z
    X-VFTNWL-Z
    X-DVFTNWL-Z
    X-WDVFTNWL-Z
    X-SWDVFTNWL-Z
    X-NSWDVFTNWL-Z
    X-LNSWDVFTNWL-Z
    X-KLNSWDVFTNWL-Z
    X-QKLNSWDVFTNWL-Z
    X-LQKLNSWDVFTNWL-Z
    X-ELQKLNSWDVFTNWL-Z
    X-YELQKLNSWDVFTNWL-Z
    X-MYELQKLNSWDVFTNWL-Z
    X-NMYELQKLNSWDVFTNWL-Z
    X-KNMYELQKLNSWDVFTNWL-Z
    X-EKNMYELQKLNSWDVFTNWL-Z
    X-QEKNMYELQKLNSWDVFTNWL-Z
    X-QQEKNMYELQKLNSWDVFTNWL-Z
    X-IQQEKNMYELQKLNSWDVFTNWL-Z
    X-QIQQEKNMYELQKLNSWDVFTNWL-Z
    X-AQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-QAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-EQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-LEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-SLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-QSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-SQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-ISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-NISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-ANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-EANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    X-LEANISQSLEQAQIQQEKNMYELQKLNSWDVFTNWL-Z
    The one letter amino acid code is used.
    Additionally, “X” may represent an amino group, a hydrophobic group, including but not limited to carbobenzoxyl, dansyl, or T-butyloxycarbonyl; an acetyl group; a 9-fluorenylmethoxy-carbonyl (FMOC) group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
    “Z” may represent a carboxyl group; an amido group; a T-butyloxycarbonyl group; a macromolecular carrier group including but not limited to lipid-fatty acid conjugates, polyethylene glycol, or carbohydrates.
  • DP178 and DP107 analogs are recognized or identified, for example, by utilizing one or more of the 107×178×4, ALLMOTI5 or PLZIP computer-assisted search strategies described and demonstrated, below, in the Examples presented in Sections 9 through 16 and 19 through 25. The search strategy identifies additional peptide regions which are predicted to have structural and/or amino acid sequence features similar to those of DP107 and/or DP178.
  • The search strategies are described fully, below, in the Example presented in Section 9. While this search strategy is based, in part, on a primary amino acid motif deduced from DP107 and DP178, it is not based solely on searching for primary amino acid sequence homologies, as such protein sequence homologies exist within, but not between major groups of viruses. For example, primary amino acid sequence homology is high within the TM protein of different strains of HIV-1 or within the TM protein of different isolates of simian immunodeficiency virus (SIV). Primary amino acid sequence homology between HIV-1 and SIV, however, is low enough so as not to be useful. It is not possible, therefore, to find peptide regions similar to DP107 or DP178 within other viruses, or within non-viral organisms, whether structurally, or otherwise, based on primary sequence homology, alone.
  • Further, while it would be potentially useful to identify primary sequence arrangements of amino acids based on, for example, the physical chemical characteristics of different classes of amino acids rather than based on the specific amino acids themselves, such search strategies have, until now, proven inadequate. For example, a computer algorithm designed by Lupas et al. to identify coiled-coil propensities of regions within proteins (Lupas, A., et al., 1991 Science 252:1162-1164) is inadequate for identifying protein regions analogous to DP107 or DP178.
  • Specifically, analysis of HIV-1 gp160 (containing both gp120 and gp41) using the Lupas algorithm does not identify the coiled-coil region within DP107. It does, however, identify a region within DP178 beginning eight amino acids N-terminal to the start of DP178 and ending eight amino acids from the C-terminus. The DP107 peptide has been shown experimentally to form a stable coiled coil. A search based on the Lupas search algorithm, therefore, would not have identified the DP107 coiled-coil region. Conversely, the Lupas algorithm identified the DP178 region as a potential coiled-coil motif. However, the peptide derived from the DP178 region failed to form a coiled coil in solution.
  • A possible explanation for the inability of the Lupas search algorithm to accurately identify coiled-coil sequences within the HIV-1 TM, is that the Lupas algorithm is based on the structure of coiled coils from proteins that are not structurally or functionally similar to the TM proteins of viruses, antiviral peptides (e.g. DP107 and DP178) of which are an object of this invention.
  • The computer search strategy of the invention, as demonstrated in the Examples presented below, in Sections 9 through 16 and 19 through 25, successfully identifies regions of proteins similar to DP107 or DP178. This search strategy was designed to be used with a commercially-available sequence database package, preferably PC/Gene.
  • A series of search motifs, the 107×178×4, ALLMOTI5 and PLZIP motifs, were designed and engineered to range in stringency from strict to broad, as discussed in this Section and in Section 9, with 107×178×4 being preferred. The sequences identified via such search motifs, such as those listed in Tables V-XIV, below, potentially exhibit antifusogenic, such as antiviral, activity, may additionally be useful in the identification of antifusogenic, such as antiviral, compounds, and are intended to be within the scope of the invention.
  • Coiled-coiled sequences are thought to consist of heptad amino acid repeats. For ease of description, the amino acid positions within the heptad repeats are sometimes referred to as A through G, with the first position being A, the second B, etc. The motifs used to identify DP107-like and DP178-like sequences herein are designed to specifically search for and identify such heptad repeats. In the descriptions of each of the motifs described, below, amino acids enclosed by brackets, i.e., [ ], designate the only amino acid residues that are acceptable at the given position, while amino acids enclosed by braces, i.e., { }, designate the only amino acids which are unacceptable at the given heptad position. When a set of bracketed or braced amino acids is followed by a number in parentheses i.e., ( ), it refers to the number of subsequent amino acid positions for which the designated set of amino acids hold, e.g, a (2) means “for the next two heptad amino acid positions”.
      • The ALLMOTI5 is written as follows:
      • {CDGHP}-{CFP}(2)-{CDGHP}-{CFP}(3)-
      • {CDGHP}-{CFP}(2)-{CDGHP}-{CFP}(3)-
      • {CDGHP}-{CFP}(2)-{CDGHP}-{CFP}(3)-
      • {CDGHP}-{CFP}(2)-{CDGHP}-{CFP}(3)-
      • {CDGHP}-{CFP}(2)-{CDGHP}-{CFP}(3)-
  • Translating this motif, it would read: “at the first (A) position of the heptad, any amino acid residue except C, D, G, H, or P is acceptable, at the next two (B,C) amino acid positions, any amino acid residue except C, F, or P is acceptable, at the fourth heptad position (D), any amino acid residue except C, D, G, H, or P is acceptable, at the next three (E, F, G) amino acid positions, any amino acid residue except C, F, or P is acceptable. This motif is designed to search for five consecutive heptad repeats (thus the repeat of the first line five times), meaning that it searches for 35-mer sized peptides. It may also be designed to search for 28-mers, by only repeating the initial motif four times. With respect to the ALLMOTI5 motif, a 35-mer search is preferred. Those viral (non-bacteriophage) sequences identified via such an ALLMOTI5 motif are listed in Table V, below, at the end of this Section. The viral sequences listed in Table V potentially exhibit antiviral activity, may be useful in the identification of antiviral compounds, and are intended to be within the scope of the invention. In those instances wherein a single gene exhibits greater than one sequence recognized by the ALLMOTI5 search motif, the amino a cid residue numbers of these sequences are listed under “Area 2”, Area 3”, etc. This convention is used for each of the Tables listed, below, at the end of this Section.
  • The 107×178×4 motif is written as follows:
      • [EFIKLNQSTVWY]-{CFMP}(2)-[EFIKLNQSTVWY]-{CFMP}(3)-
      • [EFIKLNQSTVWY]-{CFMP}(2)-[EFIKLNQSTVWY]-{CFMP}(3)-
      • [EFIKLNQSTVWY]-{CFMP}(2)-[EFIKLNQSTVWY]-{CFMP}(3)-
      • [EFIKLNQSTVWY]-{CFMP}(2)-[EFIKLNQSTVWY]-{CFMP}(3)-
  • Translating this motif, it would read: “at the first (A) position of the heptad, only amino acid residue E, F, I, K, L, N, Q, S, T, V, W, or Y is acceptable, at the next two (B,C) amino acid positions, any amino acid residue except C, F, M or P is acceptable, at the fourth position (D), only amino acid residue E, F, I, K, L, N, Q, S, T, V, W, or Y is acceptable, at the next three (E, F, G) amino acid positions, any amino acid residue except C, F, M or P is acceptable. This motif is designed to search for four consecutive heptad repeats (thus the repeat of the first line four times), meaning that it searches for 28-mer sized peptides. It may also be designed to search for 35-mers, by repeating the initial motif five times. With respect to the 107×178×4 motif, a 28-mer search is preferred.
  • Those viral (non-bacteriophage) sequences identified via such a 107×178×4 motif are listed in Table VI, below, at the end of this Section, with those viral (non-bacteriophage) sequences listed in Table VII, below at the end of this Section, being preferred.
  • The 107×178×4 search motif was also utilized to identify non-viral procaryotic protein sequences, as listed in Table VIII, below, at the end of this Section. Further, this search motif was used to reveal a number of human proteins. The results of this human protein 107×178×4 search is listed in Table IX, below, at the end of this Section. The sequences listed in Tables VIII and IX, therefore, reveal peptides which may be useful as antifusogenic compounds or in the identification of antifusogenic compounds, and are intended to be within the scope of the invention.
  • The PLZIP series of motifs are as listed in FIG. 19. These motifs are designed to identify leucine zipper coiled-coil like heptads wherein at least one proline residue is present at some predefined distance N-terminal to the repeat. These PLZIP motifs find regions of proteins with similarities to HIV-1 DP178 generally located just N-terminal to the transmembrane anchor. These motifs may be translated according to the same convention described above. Each line depicted in FIG. 19 represents a single, complete search motif. “X” in these motifs refers to any amino acid residue. In instances wherein a motif contains two numbers within parentheses, this refers to a variable number of amino acid residues. For example, X (1,12) is translated to “the next one to twelve amino acid residues, inclusive, may be any amino acid”.
  • Tables X through XIV, below, at the end of this Section, list sequences identified via searches conducted with such PLZIP motifs. Specifically, Table X lists viral sequences identified via PCTLZIP, P1CTLZIP and P2CTLZIP search motifs, Table XI lists viral sequences identified via P3CTLZIP, P4CTLZIP, P5CTLZIP and P6CTLZIP search motifs, Table XII lsts viral sequences identified via P7CTLZIP, P8CTLZIP and P9CTLZIP search motifs, Table XIII lists viral sequences identified via P12LZIPC searches and Table XIV lists viral sequences identified via P23TLZIPC search motifs The viral sequences listed in these tables represent peptides which potentially exhibit antiviral activity, may be useful in the identification of antiviral compounds, and are intended to be within the scope of the invention.
  • The Examples presented in Sections 17, 18, 26 and 27 below, demonstrate that viral sequences identified via the motif searches described herein identify substantial antiviral characteristics. Specifically, the Example presented in Section 17 describes peptides with anti-respiratory syncytial virus activity, the Example presented in Section 18 describes peptides with anti-parainfluenza virus activity, the Example presented in Section 26 describes peptides with anti-measles virus activity and the Example presented in Section 27 describes peptides with anti-simian immunodeficiency virus activity.
  • The DP107 and DP178 analogs may, further, contain any of the additional groups described for DP178, above, in Section 5.1. For example, these peptides may include any of the additional amino-terminal groups as described above for “X” groups, and may also include any of the carboxy-terminal groups as described, above, for “Z” groups.
  • Additionally, truncations of the identified DP107 and DP178 peptides are among the peptides of the invention. Further, such DP107 and DP178 analogs and DP107/DP178 analog truncations may exhibit one or more amino acid substitutions, insertion, and/or deletions. The DP178 analog amino acid substitutions, insertions and deletions, are as described, above, for DP178-like peptides in Section 5.1. The DP-107 analog amino acid substitutions, insertions and deletions are also as described, above, for DP107-like peptides in Section 5.2.
  • Tables XV through XXII, below, present representative examples of such DP107/DP178 truncations. Specifically, Table XV presents Respiratory Syncytial Virus F1 region DP107 analog carboxy truncations, Table XVI presents Respiratory Syncytial Virus F1 region DP107 analog amino truncations, Table XVII presents Respiratory Syncytial Virus F1 region DP178 analog carboxy truncations, Table XVIII presents Respiratory Syncytial Virus F1 region DP178 analog amino truncations, Table XIX presents Human Parainfluenza Virus 3 F1 region DP178 analog carboxy truncations, Table XX presents Human Parainfluenza Virus 3 F1 region DP178 analog amino truncations, Table XXI presents Human Parainfluenza Virus 3 F1 region DP107 analog carboxy truncations and Table XXII presents Human Parainfluenza Virus 3 F1 region DP107 analog amino truncations. Further, Table XXIII, below, presents DP107/DP178 analogs and analog truncations which exhibit substantial antiviral activity. These antiviral peptides are grouped according to the specific virus which they inhibit, including respiratory syncytial virus, human parainfluenza virus 3, simian immunodeficiency virus and measles virus.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00001
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00002
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00003
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00004
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00005
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00006
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00007
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00008
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00009
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00010
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00011
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00012
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00013
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00014
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00015
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00016
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00017
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00018
    Please refer to the end of the specification for access instructions.
  • Lengthy table referenced here
    US20100291680A1-20101118-T00019
    Please refer to the end of the specification for access instructions.
  • 6.1.3 Synthesis of Peptides
  • The peptides of the invention may be synthesized or prepared by techniques well known in the art. See, for example, Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman and Co., NY, which is incorporated herein by reference in its entirety. Short peptides, for example, can be synthesized on a solid support or in solution. Longer peptides may be made using recombinant DNA techniques. Here, the nucleotide sequences encoding the peptides of the invention may be synthesized, and/or cloned, and expressed according to techniques well known to those of ordinary skill in the art. See, for example, Sambrook, et al., 1989, Molecular Cloning, A Laboratory Manual, Vols. 1-3, Cold Spring Harbor Press, NY.
  • The peptides of the invention may alternatively be synthesized such that one or more of the bonds which link the amino acid residues of the peptides are non-peptide bonds. These alternative non-peptide bonds may be formed by utilizing reactions well known to those in the art, and may include, but are not limited to imino, ester, hydrazide, semicarbazide, and azo bonds, to name but a few. In yet another embodiment of the invention, peptides comprising the sequences described above may be synthesized with additional chemical groups present at their amino and/or carboxy termini, such that, for example, the stability, bioavailability, and/or inhibitory activity of the peptides is enhanced. For example, hydrophobic groups such as carbobenzoxyl, dansyl, or t-butyloxycarbonyl groups, may be added to the peptides' amino termini. Likewise, an acetyl group or a 9-fluorenylmethoxy-carbonyl group may be placed at the peptides' amino termini. (See “X” in Tables I to IV, above.) Additionally, the hydrophobic group, t-butyloxycarbonyl, or an amido group may be added to the peptides' carboxy termini. (See “Z” in Tables I to IV, above.)
  • Further, the peptides of the invention may be synthesized such that their steric configuration is altered. For example, the D-isomer of one or more of the amino acid residues of the peptide may be used, rather than the usual L-isomer.
  • Still further, at least one of the amino acid residues of the peptides of the invention may be substituted by one of the well known non-naturally occurring amino acid residues. Alterations such as these may serve to increase the stability, bioavailability and/or inhibitory action of the peptides of the invention.
  • Any of the peptides described above may, additionally, have a macromolecular carrier group covalently attached to their amino and/or carboxy termini. Such macromolecular carrier groups may include, for example, lipid-fatty acid conjugates, polyethylene glycol, carbohydrates or additional peptides. “X”, in Tables I to IV, above, may therefore additionally represent any of the above macromolecular carrier groups covalently attached to the amino terminus of a peptide, with an additional peptide group being preferred. Likewise, “Z”, in Tables I to IV, may additionally represent any of the macromolecular carrier groups described above.
  • 6.1.4 Assays for Anti-Membrane Fusion Activity
  • Described herein, are methods for ability of a compound, such as the peptides of the invention, to inhibit membrane fusion events. Specifically, assays for cell fusion events are described in Section 5.5.1, below, and assays for antiviral activity are described in Section 5.5.2, below.
  • 6.1.4.1 Assays for Cell Fusion Events
  • Assays for cell fusion events are well known to those of skill in the art, and may be used in conjunction, for example, with the peptides of the invention to test the peptides' antifusogenic capabilities.
  • Cell fusion assays are generally performed in vitro. Such an assay may comprise culturing cells which, in the absence of any treatment would undergo an observable level of syncytial formation. For example, uninfected cells may be incubated in the presence of cells chronically infected with a virus that induces cell fusion. Such viruses may include, but are not limited to, HIV, SIV, or respiratory syncytial virus.
  • For the assay, cells are incubated in the presence of a peptide to be assayed. For each peptide, a range of peptide concentrations may be tested. This range should include a control culture wherein no peptide has been added.
  • Standard conditions for culturing cells, well known to those of ordinary skill in the art, are used. After incubation for an appropriate period (24 hours at 37° C., for example) the culture is examined microscopically for the presence of multinucleated giant cells, which are indicative of cell fusion and syncytial formation. Well known stains, such as crystal violet stain, may be used to facilitate the visualization of syncytial formation.
  • 6.1.4.2 Assays for Antiviral Activity
  • The antiviral activity exhibited by the peptides of the invention may be measured, for example, by easily performed in vitro assays, such as those described below, which can test the peptides' ability to inhibit syncytia formation, or their ability to inhibit infection by cell-free virus. Using these assays, such parameters as the relative antiviral activity of the peptides, exhibit against a given strain of virus and/or the strain specific inhibitory activity of the peptide can be determined.
  • A cell fusion assay may be utilized to test the peptides' ability to inhibit viral-induced, such as HIV-induced, syncytia formation in vitro. Such an assay may comprise culturing uninfected cells in the presence of cells chronically infected with a syncytial-inducing virus and a peptide to be assayed. For each peptide, a range of peptide concentrations may be tested. This range should include a control culture wherein no peptide has been added. Standard conditions for culturing, well known to those of ordinary skill in the art, are used. After incubation for an appropriate period (24 hours at 37° C., for example) the culture is examined microscopically for the presence of multinucleated giant cells, which are indicative of cell fusion and syncytia formation. Well known stains, such as crystal violet stain, may be used to facilitate syncytial visualization. Taking HIV as an example, such an assay would comprise CD-4+ cells (such as Molt or CEM cells, for example) cultured in the presence of chronically HIV-infected cells and a peptide to be assayed.
  • Other well known characteristics of viral infection may also be assayed to test a peptide's antiviral capabilities. Once again taking HIV as an example, a reverse transcriptase (RT) assay may be utilized to test the peptides' ability to inhibit infection of CD-4+ cells by cell-free HIV. Such an assay may comprise culturing an appropriate concentration (i.e., TCID50) of virus and CD-4+ cells in the presence of the peptide to be tested. Culture conditions well known to those in the art are used. As above, a range of peptide concentrations may be used, in addition to a control culture wherein no peptide has been added. After incubation for an appropriate period (e.g., 7 days) of culturing, a cell-free supernatant is prepared, using standard procedures, and tested for the present of RT activity as a measure of successful infection. The RT activity may be tested using standard techniques such as those described by, for example, Goff et al. (Goff, S. et al., 1981, J. Virol. 38:239-248) and/or Willey et al. (Willey, R. et al., 1988, J. Virol. 62:139-147). These references are incorporated herein by reference in their entirety.
  • Standard methods which are well-known to those of skill in the art may be utilized for assaying non-retroviral activity. See, for example, Pringle et al. (Pringle, C. R. et al., 1985, J. Medical Virology 17:377-386) for a discussion of respiratory syncytial virus and parainfluenza virus activity assay techniques. Further, see, for example, “Zinsser Microbiology”, 1988, Joklik, W. K. et al., eds., Appleton & Lange, Norwalk, Conn., 19th ed., for a general review of such techniques. These references are incorporated by reference herein in their entirety. In addition, the Examples presented below, in Sections 17, 18, 26 and 27 each provide additional assays for the testing of a compound's antiviral capability.
  • In vivo assays may also be utilized to test, for example, the antiviral activity of the peptides of the invention. To test for anti-HIV activity, for example, the in vivo model described in Barnett et al. (Barnett, S. W. et al., 1994, Science 266:642-646) may be used.
  • Additionally, anti-RSV activity can be assayed in vivo via well known mouse models. For example, RSV can be administered intranasally to mice of various inbred strains. Virus replicates in lungs of all strains, but the highest titers are obtained in P/N, C57L/N and DBA/2N mice. Infection of BALB/c mice produces an asymptomatic bronchiolitis characterized by lymphocytic infiltrates and pulmonary virus titers of 104 to 105 pfu/g of lung tissue (Taylor, G. et al., 1984, Infect. Immun. 43:649-655).
  • Cotton rat models of RSV are also well known. Virus replicates to high titer in the nose and lungs of the cotton rat but produces few if any signs of inflammation.
  • 6.1.5 Uses of the Peptides of the Invention
  • The peptides of the invention may be utilized as antifusogenic or antiviral compounds, or as compounds which modulate intracellular processes involving coiled coil peptide structures. Further, such peptides may be used to identify agents which exhibit antifusogenic, antiviral or intracellular modulatory activity. Still further, the peptides of the invention may be utilized as organism or viral type/subtype-specific diagnostic tools.
  • The antifusogenic capability of the peptides of the invention may additionally be utilized to inhibit or treat/ameliorate symptoms caused by processes involving membrane fusion events. Such events may include, for example, virus transmission via cell-cell fusion, abnormal neurotransmitter exchange via cell-fusion, and sperm-egg fusion. Further, the peptides of the invention may be used to inhibit free viral, such as retroviral, particularly HIV, transmission to uninfected cells wherein such viral infection involves membrane fusion events or involves fusion of a viral structure with a cell membrane. Among the intracellular disorders involving coiled coil peptides structures which may be ameliorated by the peptides of the invention are disorders involving, for example, bacterial toxins.
  • With respect to antiviral activity, the viruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to all strains of the viruses listed above, in Tables V through VII, and IX through XIV.
  • These viruses include, for example, human retroviruses, particularly HIV-1 and HIV-2 and the human T-lymphocyte viruses (HTLV-I and II). The non-human retroviruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to bovine leukosis virus, feline sarcoma and leukemia viruses, simian immunodeficiency, sarcoma and leukemia viruses, and sheep progress pneumonia viruses.
  • Non retroviral viruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to human respiratory syncytial virus, canine distemper virus, newcastle disease virus, human parainfluenza virus, influenza viruses, measles viruses, Epstein-Barr viruses, hepatitis B viruses, and simian Mason-Pfizer viruses.
  • Non enveloped viruses whose transmission may be inhibited by the peptides of the invention include, but are not limited to picornaviruses such as polio viruses, hepatitis A virus, enterovirus, echoviruses and coxsackie viruses, papovaviruses such as papilloma virus, parvoviruses, adenoviruses and reoviruses.
  • As discussed more fully, below, in Section 5.5.1 and in the Example presented, below, in Section 8, DP107, DP178, DP107 analog and DP178 analog peptides form non-covalent protein-protein interactions which are required for normal activity of the virus. Thus, the peptides of the invention may also be utilized as components in assays for the identification of compounds that interfere with such protein-protein interactions and may, therefore, act as antiviral agents. These assays are discussed, below, in Section 5.5.1.
  • As demonstrated in the Example presented below in Section 6, the antiviral activity of the peptides of the invention may show a pronounced type and subtype specificity, i.e., specific peptides may be effective in inhibiting the activity of only specific viruses. This feature of the invention presents many advantages. One such advantage, for example, lies in the field of diagnostics, wherein one can use the antiviral specificity of the peptide of the invention to ascertain the identity of a viral isolate. With respect to HIV, one may easily determine whether a viral isolate consists of an HIV-1 or HIV-2 strain. For example, uninfected CD-4+ cells may be co-infected with an isolate which has been identified as containing HIV the DP178 (SEQ ID NO:1) peptide, after which the retroviral activity of cell supernatants may be assayed, using, for example, the techniques described above in Section 5.2. Those isolates whose retroviral activity is completely or nearly completely inhibited contain HIV-1. Those isolates whose viral activity is unchanged or only reduced by a small amount, may be considered to not contain HIV-1. Such an isolate may then be treated with one or more of the other DP178 peptides of the invention, and subsequently be tested for its viral activity in order to determine the identify of the viral isolate. The DP107 and DP178 analogs of the invention may also be utilized in a diagnostic capacity specific to the type and subtype of virus or organism in which the specific peptide sequence is found. A diagnostic procedure as described, above, for DP178, may be used in conjunction with the DP107/DP178 analog of interest.
  • 6.1.5.1 Screening Assays
  • As demonstrated in the Example presented in Section 8, below, DP107 and DP178 portions of the TM protein gp41 form non-covalent protein-protein interactions. As is also demonstrated, the maintenance of such interactions is necessary for normal viral infectivity. Thus, compounds which bind DP107, bind DP178, and/or act to disrupt normal DP107/DP178 protein-protein interactions may act as antifusogenic, antiviral or cellular modulatory agents. Described below are assays for the identification of such compounds. Note that, while, for ease and clarity of discussion, DP107 and DP178 peptides will be used as components of the assays described, but it is to be understood that any of the DP107 analog or DP178 analog peptides described, above, in Sections 5.1 through 5.3 may also be utilized as part of these screens for compounds.
  • Compounds which may be tested for an ability to bind DP107, DP178, and/or disrupt DP107/DP178 interactions, and which therefore, potentially represent antifusogenic, antiviral or intracellular modulatory compounds, include, but are not limited to, peptides made of D- and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam, K. S. et al., 1991, Nature 354:82-84), phosphopeptides (in, for example, the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang, Z. et al., 1993, Cell 72:767-778), antibodies, and small organic or inorganic molecules. Synthetic compounds, natural products, and other sources of potentially effective materials may be screened in a variety of ways, as described in this Section.
  • The compounds, antibodies, or other molecules identified may be tested, for example, for an ability to inhibit cell fusion or viral activity, utilizing, for example, assays such as those described, above, in Section 5.5.
  • Among the peptides which may be tested are soluble peptides comprising DP107 and/or DP178 domains, and peptides comprising DP107 and/or DP178 domains having one or more mutations within one or both of the domains, such as the M41-P peptide described, below, in the Example presented in Section 8, which contains a isoleucine to proline mutation within the DP178 sequence.
  • In one embodiment of such screening methods is a method for identifying a compound to be tested for antiviral ability comprising:
      • (a) exposing at least one compound to a peptide comprising a DP107 peptide for a time sufficient to allow binding of the compound to the DP107 peptide;
      • (b) removing non-bound compounds; and
      • (c) determining the presence of the compound bound to the DP107 peptide,
        thereby identifying an agent to be tested for antiviral ability.
  • In a second embodiment of such screening methods is a method for identifying a compound to be tested for antiviral ability comprising:
      • (a) exposing at least one compound to a peptide comprising a DP178 peptide for a time sufficient to allow binding of the compound to the DP178 peptide;
      • (b) removing non-bound compounds; and
      • (c) determining the presence of the compound bound to the DP178 peptide,
        thereby identifying an agent to be tested for antiviral ability.
  • One method utilizing these types of approaches that may be pursued in the isolation of such DP107-binding or DP178-binding compounds is an assay which would include the attachment of either the DP107 or the DP178 peptide to a solid matrix, such as, for example, agarose or plastic beads, microtiter plate wells, petri dishes, or membranes composed of for example, nylon or nitrocellulose. In such an assay system, either the DP107 or DP178 protein may be anchored onto a solid surface, and the compound, or test substance, which is not anchored, is labeled, either directly or indirectly. In practice, microtiter plates are conveniently utilized. The anchored component may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the protein and drying.
  • Alternatively, an immobilized antibody, preferably a monoclonal antibody, specific for the protein may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.
  • In order to conduct the assay, the labeled compound is added to the coated surface containing the anchored DP107 or DP178 peptide. After the reaction is complete, unreacted components are removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the compound is pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the labeled component is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the compound (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).
  • Alternatively, such an assay can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for DP107 or DP178, whichever is appropriate for the given assay, or ab antibody specific for the compound, i.e., the test substance, in order to anchor any complexes formed in solution, and a labeled antibody specific for the other member of the complex to detect anchored complexes.
  • By utilizing procedures such as this, large numbers of types of molecules may be simultaneously screened for DP107 or DP178-binding capability, and thus potential antiviral activity.
  • Further, compounds may be screened for an ability to inhibit the formation of or, alternatively, disrupt DP107/DP178 complexes. Such compounds may then be tested for antifusogenic, antiviral or intercellular modulatory capability. For ease of description, DP107 and DP178 will be referred to as “binding partners.” Compounds that disrupt such interactions may exhibit antiviral activity. Such compounds may include, but are not limited to molecules such as antibodies, peptides, and the like described above.
  • The basic principle of the assay systems used to identify compounds that interfere with the interaction between the DP107 and DP178 peptides involves preparing a reaction mixture containing peptides under conditions and for a time sufficient to allow the two peptides to interact and bind, thus forming a complex. In order to test a compound for disruptive activity, the reaction is conducted in the presence and absence of the test compound, i.e., the test compound may be initially included in the reaction mixture, or added at a time subsequent to the addition of one of the binding partners; controls are incubated without the test compound or with a placebo. The formation of any complexes between the binding partners is then detected. The formation of a complex in the control reaction, but not in the reaction mixture containing the test compound indicates that the compound interferes with the interaction of the DP107 and DP178 peptides.
  • The assay for compounds that interfere with the interaction of the binding partners can be conducted in a heterogeneous or homogeneous format. Heterogeneous assays involve anchoring one of the binding partners onto a solid phase and detecting complexes anchored on the solid phase at the end of the reaction. In homogeneous assays, the entire reaction is carried out in a liquid phase. In either approach, the order of addition of reactants can be varied to obtain different information about the compounds being tested. For example, test compounds that interfere with the interaction between the binding partners, e.g., by competition, can be identified by conducting the reaction in the presence of the test substance; i.e., by adding the test substance to the reaction mixture prior to or simultaneously with the binding partners. On the other hand, test compounds that disrupt preformed complexes, e.g. compounds with higher binding constants that displace one of the binding partners from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed. The various formats are described briefly below.
  • In a heterogeneous assay system, one binding partner, e.g., either the DP107 or DP178 peptide, is anchored onto a solid surface, and its binding partner, which is not anchored, is labeled, either directly or indirectly. In practice, microtiter plates are conveniently utilized. The anchored species may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished simply by coating the solid surface with a solution of the protein and drying. Alternatively, an immobilized antibody specific for the protein may be used to anchor the protein to the solid surface. The surfaces may be prepared in advance and stored.
  • In order to conduct the assay, the binding partner of the immobilized species is added to the coated surface with or without the test compound. After the reaction is complete, unreacted components are removed (e.g., by washing) and any complexes formed will remain immobilized on the solid surface. The detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the binding partner was pre-labeled, the detection of label immobilized on the surface indicates that complexes were formed. Where the binding partner is not pre-labeled, an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the binding partner (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody). Depending upon the order of addition of reaction components, test compounds which inhibit complex formation or which disrupt preformed complexes can be detected.
  • Alternatively, the reaction can be conducted in a liquid phase in the presence or absence of the test compound, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for one binding partner to anchor any complexes formed in solution, and a labeled antibody specific for the other binding partner to detect anchored complexes. Again, depending upon the order of addition of reactants to the liquid phase, test compounds which inhibit complex or which disrupt preformed complexes can be identified.
  • In an alternate embodiment of the invention, a homogeneous assay can be used. In this approach, a preformed complex of the DP107 and DP178 peptides is prepared in which one of the binding partners is labeled, but the signal generated by the label is quenched due to complex formation (see, e.g., U.S. Pat. No. 4,109,496 by Rubenstein which utilizes this approach for immunoassays). The addition of a test substance that competes with and displaces one of the binding partners from the preformed complex will result in the generation of a signal above background. In this way, test substances which disrupt DP-107/DP-178 protein-protein interaction can be identified.
  • In an alternative screening assay, test compounds may be assayed for the their ability to disrupt a DP178/DP107 interaction, as measured immunometrically using an antibody specifically reactive to a DP107/DP178 complex (i.e., an antibody that recognizes neither DP107 nor DP178 individually). Such an assay acts as a competition assay, and is based on techniques well known to those of skill in the art.
  • The above competition assay may be described, by way of example, and not by way of limitation, by using the DP178 and M41Δ178 peptides and by assaying test compounds for the disruption of the complexes formed by these two peptides by immunometrically visualizing DP178/M41Δ178 complexes via the human recombinant Fab, Fab-d, as described, below, in the Example presented in Section 8. M41Δ178 is a maltose binding fusion protein containing a gp41 region having its DP178 domain deleted, and is described, below, in the Example presented in Section 8.
  • Utilizing such an assay, M41Δ178 may be immobilized onto solid supports such as microtiter wells. A series of dilutions of a test compound may then be added to each M41Δ178-containing well in the presence of a constant concentration of DP-178 peptide. After incubation, at, for example, room temperature for one hour, unbound DP-178 and test compound are removed from the wells and wells are then incubated with the DP178/M41Δ178-specific Fab-d antibody. After incubation and washing, unbound Fab-d is removed from the plates and bound Fab-d is quantitated. A no-inhibitor control should also be conducted. Test compounds showing an ability to disrupt DP178/M41Δ178 complex formation are identified by their concentration-dependent decrease in the level of Fab-d binding.
  • A variation of such an assay may be utilized to perform a rapid, high-throughput binding assay which is capable of directly measuring DP178 binding to M41Δ178 for the determination of binding constants of the ligand of inhibitory constants for competitors of DP178 binding.
  • Such an assay takes advantage of accepted radioligand and receptor binding principles. (See, for example, Yamamura, H. I. et al., 1985, “Neurotransmitter Receptor Binding”, 2nd ed., Raven Press, NY.) As above, M41Δ178 is immobilized onto a solid support such as a microtiter well. DP178 binding to M41Δ178 is then quantitated by measuring the fraction of DP178 that is bound as 125I-DP178 and calculating the total amount bound using a value for specific activity (dpm/μg peptide) determined for each labeled DP178 preparation. Specific binding to M41Δ178 is defined as the difference of the binding of the labeled DP178 preparation in the microtiter wells (totals) and the binding in identical wells containing, in addition, excess unlabeled DP178 (nonspecifics).
  • 6.1.6 Pharmaceutical Formulations, Dosages and Modes of Administration
  • The peptides of the invention may be administered using techniques well known to those in the art. Preferably, agents are formulated and administered systemically. Techniques for formulation and administration may be found in “Remington's Pharmaceutical Sciences”, 18th ed., 1990, Mack Publishing Co., Easton, Pa. Suitable routes may include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as, intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections, just to name a few. For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer. For such transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • In instances wherein intracellular administration of the peptides of the invention or other inhibitory agents is preferred, techniques well known to those of ordinary skill in the art may be utilized. For example, such agents may be encapsulated into liposomes, then administered as described above. Liposomes are spherical lipid bilayers with aqueous interiors. All molecules present in an aqueous solution at the time of liposome formation are incorporated into the aqueous interior. The liposomal contents are both protected from the external microenvironment and, because liposomes fuse with cell membranes, are effectively delivered into the cell cytoplasm. Additionally, due to their hydrophobicity, when small molecules are to be administered, direct intracellular administration may be achieved.
  • Nucleotide sequences encoding the peptides of the invention which are to be intracellularly administered may be expressed in cells of interest, using techniques well known to those of skill in the art. For example, expression vectors derived from viruses such as retroviruses, vaccinia viruses, adeno-associated viruses, herpes viruses, or bovine papilloma viruses, may be used for delivery and expression of such nucleotide sequences into the targeted cell population. Methods for the construction of such vectors and expression constructs are well known. See, for example, Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor N.Y., and Ausubel et al., 1989, Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.
  • With respect to HIV, peptides of the invention, particularly DP107 and DP178, may be used as therapeutics in the treatment of AIDS. In addition, the peptides may be used as prophylactic measures in previously uninfected individuals after acute exposure to an HIV virus. Examples of such prophylactic use of the peptides may include, but are not limited to, prevention of virus transmission from mother to infant and other settings where the likelihood of HIV transmission exists, such as, for example, accidents in health care settings wherein workers are exposed to HIV-containing blood products. The successful use of such treatments do not rely upon the generation of a host immune response directed against such peptides.
  • Effective dosages of the peptides of the invention to be administered may be determined through procedures well known to those in the art which address such parameters as biological half-life, bioavailability, and toxicity. Given the data presented below in Section 6, DP178, for example, may prove efficacious in vivo at doses required to achieve circulating levels of about 1 to about 10 ng per ml of peptide.
  • A therapeutically effective dose refers to that amount of the compound sufficient to result in amelioration of symptoms or a prolongation of survival in a patient. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds which exhibit large therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (e.g., the concentration of the test compound which achieves a half-maximal inhibition of the fusogenic event, such as a half-maximal inhibition of viral infection relative to the amount of the event in the absence of the test compound) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography (HPLC).
  • The peptides of the invention may, further, serve the role of a prophylactic vaccine, wherein the host raises antibodies against the peptides of the invention, which then serve to neutralize HIV viruses by, for example, inhibiting further HIV infection.
  • Administration of the peptides of the invention as a prophylactic vaccine, therefore, would comprise administering to a host a concentration of peptides effective in raising an immune response which is sufficient to neutralize HIV, by, for example, inhibiting HIV ability to infect cells. The exact concentration will depend upon the specific peptide to be administered, but may be determined by using standard techniques for assaying the development of an immune response which are well known to those of ordinary skill in the art. The peptides to be used as vaccines are usually administered intramuscularly.
  • The peptides may be formulated with a suitable adjuvant in order to enhance the immunological response. Such adjuvants may include, but are not limited to mineral gels such as aluminum hydroxide; surface active substances such as lysolecithin, pluronic polyols, polyanions; other peptides; oil emulsions; and potentially useful human adjuvants such as BCG and Corynebacterium parvum. Many methods may be used to introduce the vaccine formulations described here. These methods include but are not limited to oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, and intranasal routes.
  • Alternatively, an effective concentration of polyclonal or monoclonal antibodies raised against the peptides of the invention may be administered to a host so that no uninfected cells become infected by HIV. The exact concentration of such antibodies will vary according to each specific antibody preparation, but may be determined using standard techniques well known to those of ordinary skill in the art. Administration of the antibodies may be accomplished using a variety of techniques, including, but not limited to those described in this section.
  • For all such treatments described above, the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g. Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p 1).
  • It should be noted that the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity, or to organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administrated dose in the management of the oncogenic disorder of interest will vary with the severity of the condition to be treated and the route of administration. The dose and perhaps dose frequency, will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.
  • Use of pharmaceutically acceptable carriers to formulate the compounds herein disclosed for the practice of the invention into dosages suitable for systemic administration is within the scope of the invention. With proper choice of carrier and suitable manufacturing practice, the compositions of the present invention, in particular, those formulated as solutions, may be administered parenterally, such as by intravenous injection. The compounds can be formulated readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for oral administration. Such carriers enable the compounds of the invention to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • In addition to the active ingredients, these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. The preparations formulated for oral administration may be in the form of tablets, dragees, capsules, or solutions.
  • The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • Pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added.
  • 6.2. Example: DP178 (SEQ ID NO:1) is a Potent Inhibitor of HIV-1 Infection
  • In this example, DP178 (SEQ ID NO:1) is shown to be a potent inhibitor of HIV-1 mediated CD-4+ cell-cell fusion and infection by cell free virus. In the fusion assay, this peptide completely blocks virus induced syncytia formation at concentrations of from 1-10 ng/ml. In the infectivity assay the inhibitory concentration is somewhat higher, blocking infection at 90 ng/ml. It is further shown that DP178 (SEQ ID NO:1) shows that the antiviral activity of DP178 (SEQ ID NO:1) is highly specific for HIV-1. Additionally, a synthetic peptide, DP-185 (SEQ ID NO:3), representing a HIV-1-derived DP178 homolog is also found to block HIV-1-mediated syncytia formation.
  • 6.2.1 Materials and Methods 6.2.1.1 Peptide Synthesis
  • Peptides were synthesized using Fast Moc chemistry on an Applied Biosystems Model 431A peptide synthesizer. Generally, unless otherwise noted, the peptides contained amidated carboxy termini and acetylated amino termini. Amidated peptides were prepared using Rink resin (Advanced Chemtech) while peptides containing free carboxy termini were synthesized on Wang (p-alkoxy-benzyl-alcohol) resin (Bachem). First residues were double coupled to the appropriate resin and subsequent residues were single coupled. Each coupling step was followed by acetic anhydride capping. Peptides were cleaved from the resin by treatment with trifluoracetic acid (TFA) (10 ml), H2O (0.5 ml), thioanisole (0.5 ml), ethanedithiol (0.25 ml), and crystalline phenol (0.75 g). Purification was carried out by reverse phase HPLC. Approximately 50 mg samples of crude peptide were chromatographed on a Waters Delta Pak C18 column (19 mm×30 cm, 15μ spherical) with a linear gradient; H2O/acetonitrile 0.1% TFA. Lyophilized peptides were stored desiccated and peptide solutions were made in water at about 1 mg/ml. Electrospray mass spectrometry yielded the following results: DP178 (SEQ ID NO:1):4491.87 (calculated 4491.94); DP-180 (SEQ ID NO:2):4491.45 (calculated 4491.94); DP-185 (SEQ ID NO:3):not done (calculated 4546.97).
  • 6.2.1.2 Virus
  • The HIV-1LAI virus was obtained from R. Gallo (Popovic, M. et al., 1984, Science 224:497-508) and propagated in CEM cells cultured in RPMI 1640 containing 10% fetal calf serum. Supernatant from the infected CEM cells was passed through a 0.2 μm filter and the infectious titer estimated in a microinfectivity assay using the AA5 cell line to support virus replication. For this purpose, 250 of serial diluted virus was added to 75 μl AA5 cells at a concentration of 2×105/ml in a 96-well microtitre plate. Each virus dilution was tested in triplicate. Cells were cultured for eight days by addition of fresh medium every other day. On day 8 post infection, supernatant samples were tested for virus replication as evidenced by reverse transcriptase activity released to the supernatant. The TCID50 was calculated according to the Reed and Muench formula (Reed, L. J. et al., 1938, Am. J. Hyg. 27:493-497). The titer of the HIV-1LAI and HIV-1MN stocks used for these studies, as measured on the AA5 cell line, was approximately 1.4×106 and 3.8×104 TCID50/ml, respectively.
  • 6.2.1.3 Cell Fusion Assay
  • Approximately 7×104 Molt cells were incubated with 1×104 CEM cells chronically infected with the HIV-1LAI virus in 96-well plates (one-half area cluster plates; Costar, Cambridge, Mass.) in a final volume of 100 μl culture medium as previously described (Matthews, T. J. et al., 1987, Proc. Natl. Acad. Sci. USA 84: 5424-5428). Peptide inhibitors were added in a volume of 100 and the cell mixtures were incubated for 24 hr. at 37° C. At that time, multinucleated giant cells were estimated by microscopic examination at a 40× magnification which allowed visualization of the entire well in a single field.
  • 6.2.1.4 Cell Free Virus Infection Assay
  • Synthetic peptides were incubated at 37° C. with either 247 TCID50 (for experiment depicted in FIG. 2), or 62 TCID50 (for experiment depicted in FIG. 3) units of HIV-1LAI virus or 25 TCID50 units of HIV-2NIHZ and CEM CD4+ cells at peptide concentrations of 0, 0.04, 0.4, 4.0, and 40 μg/ml for 7 days. The resulting reverse transcriptase (RT) activity in counts per minute was determined using the assay described, below, in Section 6.1.5. See, Reed, L. J. et al., 1938, Am. J. Hyg. 27: 493-497 for an explanation of TCID50 calculations.
  • 6.2.1.5 Reverse Transcriptase Assay
  • The micro-reverse transcriptase (RT) assay was adapted from Goff et al. (Goff, S. et al., 1981, J. Virol. 38:239-248) and Willey et al. (Willey, R. et al., 1988, J. Virol. 62:139-147). Supernatants from virus/cell cultures are adjusted to 1% Triton-X100. A 10 μl sample of supernatant was added to 50 μl of RT cocktail in a 96-well U-bottom microtitre plate and the samples incubated at 37° C. for 90 min. The RT cocktail contained 75 mM KCl, 2 mM dithiothreitol, 5 mM MgCl2, 5 μg/ml poly A (Pharmacia, cat. No. 27-4110-01), 0.25 units/ml oligo dT (Pharmacia, cat. No. 27-7858-01), 0.05% NP40, 50 mM Tris-HCl, pH 7.8, 0.5 μM non-radioactive dTTP, and 10 μCi/ml 32P-dTTP (Amersham, cat. No. PB. 10167).
  • After the incubation period, 40 μl of reaction mixture was applied to a Schleicher and Schuell (S+S) NA45 membrane (or DE81 paper) saturated in 2×SSC buffer (0.3M NaCl and 0.003M sodium citrate) held in a S+S Minifold over one sheet of GB003 (S+S) filter paper, with partial vacuum applied. Each well of the minifold was washed four times with 200 μl 2×SSC, under full vacuum. The membrane was removed from the minifold and washed 2 more times in a pyrex dish with an excess of 2×SSC. Finally, the membrane was drained on absorbent paper, placed on Whatman #3 paper, covered with Saran wrap, and exposed to film overnight at −70° C.
  • 6.2.2 Results 6.2.2.1 Peptide Inhibition of Infected Cell-Induced Syncytia Formation
  • The initial screen for antiviral activity assayed peptides' ability to block syncytium formation induced by overnight co-cultivation of uninfected Molt4 cells with chronically HIV-1 infected CEM cells. The results of several such experiments are presented herein. In the first of these experiments, serial DP178 (SEQ ID NO:1) peptide concentrations between 10 μg/ml and 12.5 ng/ml were tested for blockade of the cell fusion process. For these experiments, CEM cells chronically infected with either HIV-1LAI, HIV-1MN, HIV-1RF, or HIV-1SF2 virus were cocultivated overnight with uninfected Molt 4 cells. The results (FIG. 4) show that DP178 (SEQ ID NO:1) afforded complete protection against each of the HIV-1 isolates down to the lowest concentration of DP178 (SEQ ID NO:1) used. For HIVLAI inhibition, the lowest concentration tested was 12.5 ng/ml; for all other HIV-1 viruses, the lowest concentration of DP178 (SEQ ID NO:1) used in this study was 100 ng/ml. A second peptide, DP-180 (SEQ ID NO:2), containing the same amino acid residues as DP178 (SEQ ID NO:1) but arranged in a random order exhibited no evidence of anti-fusogenic activity even at the high concentration of 40 μg/ml (FIG. 4). These observations indicate that the inhibitory effect of DP178 (SEQ ID NO:1) is primary sequence-specific and not related to non-specific peptide/protein interactions. The actual endpoint (i.e., the lowest effective inhibitory concentration) of DP178 inhibitory action is within the range of 1-10 ng/ml.
  • The next series of experiments involved the preparation and testing of a DP178 (SEQ ID NO:1) homolog for its ability to inhibit HIV-1-induced syncytia formation. As shown in FIG. 1, the sequence of DP-185 (SEQ ID NO:3) is slightly different from DP178 (SEQ ID NO:1) in that its primary sequence is taken from the HIV-1SF2 isolate and contains several amino acid differences relative to DP178 (SEQ ID NO:1) near the N terminus. As shown in FIG. 4, DP-185 (SEQ ID NO:3), exhibits inhibitory activity even at 312.5 ng/ml, the lowest concentration tested.
  • The next series of experiments involved a comparison of DP178 (SEQ ID NO:1) HIV-1 and HIV-2 inhibitory activity. As shown in FIG. 5, DP178 (SEQ ID NO:1) blocked HIV-1-mediated syncytia formation at peptide concentrations below 1 ng/ml. DP178 (SEQ ID NO:1) failed, however, to block HIV-2 mediated syncytia formation at concentrations as high as 10 μg/ml. This striking 4 log selectivity of DP178 (SEQ ID NO:1) as an inhibitor of HIV-1-mediated cell fusion demonstrates an unexpected HIV-1 specificity in the action of DP178 (SEQ ID NO:1). DP178 (SEQ ID NO:1) inhibition of HIV-1-mediated cell fusion, but the peptide's inability to inhibit HIV-2 medicated cell fusion in the same cell type at the concentrations tested provides further evidence for the high degree of selectivity associated with the antiviral action of DP178 (SEQ ID NO:1).
  • 6.2.2.2 Peptide Inhibition of Infection by Cell-Free Virus
  • DP178 (SEQ ID:1) was next tested for its ability to block CD-4+ CEM cell infection by cell free HIV-1 virus. The results, shown in FIG. 2, are from an experiment in which DP178 (SEQ ID NO:1) was assayed for its ability to block infection of CEM cells by an HIV-1LAI isolate. Included in the experiment were three control peptides, DP-116 (SEQ ID NO:9), DP-125 (SEQ ID NO:8), and DP-118 (SEQ ID NO:10). DP-116 (SEQ ID NO:9) represents a peptide previously shown to be inactive using this assay, and DP-125 (SEQ ID:8; Wild, C. et al., 1992, Proc. Natl. Acad. Sci. USA 89:10, 537) and DP-118 (SEQ ID NO:10) are peptides which have previously been shown to be active in this assay. Each concentration (0, 0.04, 0.4, 4, and 40 μg/ml) of peptide was incubated with 247 TCID50 units of HIV-1LAI virus and CEM cells. After 7 days of culture, cell-free supernatant was tested for the presence of RT activity as a measure of successful infection. The results, shown in FIG. 2, demonstrate that DP178 (SEQ ID NO:1) inhibited the de novo infection process mediated by the HIV-1 viral isolate at concentrations as low as 90 ng/ml (IC50=90 ng/ml). In contrast, the two positive control peptides, DP-125 (SEQ: ID NO:8) and DP-118 (SEQ ID NO:10), had over 60-fold higher IC50 concentrations of approximately 5 μg/ml.
  • In a separate experiment, the HIV-1 and HIV-2 inhibitory action of DP178 (SEQ ID:1) was tested with CEM cells and either HIV-1LAI or HIV-2NIHZ. 62 TCID50 HIV-1LAI or 25 GCID50 HIV-2NIHZ were used in these experiments, and were incubated for 7 days. As may be seen in FIG. 3, DP178 (SEQ ID NO:1) inhibited HIV-1 infection with an IC50 of about 31 ng/ml. In contrast, DP178 (SEQ ID NO:1) exhibited a much higher IC50 for HIV-2NIHZ, thus making DP178 (SEQ ID NO:1) two logs more potent as a HIV-1 inhibitor than a HIV-2 inhibitor. This finding is consistent with the results of the fusion inhibition assays described, above, in Section 6.2.1, and further supports a significant level of selectivity (i.e., for HIV-1 over HIV-2).
  • 6.3. Example: The HIV-1 Inhibitor, DP178 (SEQ ID:1) is Non-Cytotoxic
  • In this Example, the 36 amino acid synthetic peptide inhibitor DP178 (SEQ ID NO:1) is shown to be non-cytotoxic to cells in culture, even at the highest peptide concentrations (40 μg/ml) tested.
  • 6.3.1 Materials and Methods
  • Cell proliferation and toxicity assay: Approximately 3.8×105 CEM cells for each peptide concentration were incubated for 3 days at 37° C. in T25 flasks. Peptides tested were DP178 (SEQ ID NO:1) and DP-116 (SEQ ID NO:9), as described in FIG. 1. Peptides were synthesized as described, above, in Section 6.1. The concentrations of each peptide used were 0, 2.5, 10, and 40 μg/ml. Cell counts were taken at incubation times of 0, 24, 48, and 72 hours.
  • 6.3.2 Results
  • Whether the potent HIV-1 inhibitor DP178 (SEQ ID NO:1) exhibited any cytotoxic effects was assessed by assaying the peptide's effects on the proliferation and viability of cells in culture. CEM cells were incubated in the presence of varying concentrations of DP178 (SEQ ID NO:1), and DP-116 (SEQ ID NO:9), a peptide previously shown to be ineffective as a HIV inhibitor (Wild, C. et al., 1992, Proc. Natl. Acad. Sci. USA 89:10, 537-10, 541). Additionally, cells were incubated in the absence of either peptide.
  • The results of the cytotoxicity study demonstrate that DP178 (SEQ ID NO:1) exhibits no cytotoxic effects on cells in culture. As can be seen, below, in Table XXIV, even the proliferation and viability characteristics of cells cultured for 3 days in the presence of the highest concentration of DP178 (SEQ ID NO:1) tested (40 μg/ml) do not significantly differ from the DP-116 (SEQ ID NO:9) or the no-peptide controls. The cell proliferation data is also represented in graphic form in FIG. 6. As was demonstrated in the Working Example presented above in Section 6, DP178 (SEQ ID NO:1) completely inhibits HIV-1 mediated syncytia formation at peptide concentrations between 1 and 10 ng/ml, and completely inhibits cell-free viral infection at concentrations of at least 90 ng/ml. Thus, this study demonstrates that even at peptide concentrations greater than 3 log higher than the HIV inhibitory dose, DP178 (SEQ ID NO:1) exhibits no cytotoxic effects.
  • TABLE XXIV
    % Viability
    Peptide at time (hours)
    Peptide Concentration μg/ml 0 24 48 72
    DP178 40 98 97 95 97
    (SEQ ID 10 98 97 98 98
    NO: 1) 2.5 98 93 96 96
    DP116 40 98 95 98 97
    (SEQ ID 10 98 95 93 98
    NO: 9) 2.5 98 96 98 99
    No Peptide 0 98 97 99 98
  • 6.4. Example: The Interaction of DP178 and DP107
  • Soluble recombinant forms of gp41 used in the example described below provide evidence that the DP178 peptide associates with a distal site on gp41 whose interactive structure is influenced by the DP107 leucine zipper motif. A single mutation disrupting the coiled-coil structure of the leucine zipper domain transformed the soluble recombinant gp41 protein from an inactive to an active inhibitor of HIV-1 fusion. This transformation may result from liberation of the potent DP178 domain from a molecular clasp with the leucine zipper, DP107, determinant. The results also indicate that the anti-HIV activity of various gp41 derivatives (peptides and recombinant proteins) may be due to their ability to form complexes with viral gp41 and interfere with its fusogenic process.
  • 6.4.1 Materials and Methods 6.4.1.1 Construction of Fusion Proteins and GP41 Mutants
  • Construction of fusion proteins and mutants shown in FIG. 7 was accomplished as follows: the DNA sequence corresponding to the extracellular domain of gp41 (540-686) was cloned into the Xmn I site of the expression vector pMal-p2 (New England Biolab) to give M41. The gp41 sequence was amplified from pgtat (Malim et al., 1988, Nature 355: 181-183) by using polymerase chain reaction (PCR) with upstream primer 5′-ATGACGCTGACGGTACAGGCC-3′(SEQ ID NO:11) (primer A) and downstream primer 5′-TGACTAAGCTTAATACCACAGCCAATTTGTTAT-3′(SEQ ID NO:12) (primer B). M41-P was constructed by using the T7-Gen in vitro mutagenesis kit from United States Biochemicals (USB) following the supplier's instructions. The mutagenic primer (5′-GGAGCTGCTTGGGGCCCCAGAC-3′) (SEQ ID NO:13) introduces an Ile to Pro mutation in M41 at position 578. M41Δ107, from which the DP-107 region has been deleted, was made using a deletion mutagenic primer 5′-CCAAATCCCCAGGAGCTGCTCGAGCTGCACTATACCAGAC-3′(SEQ ID NO:14) (primer C) following the USB T7-Gen mutagenesis protocol. M41Δ178, from which the DP-178 region has been deleted, was made by cloning the DNA fragment corresponding to gp41 amino acids 540-642 into the Xmn I site of pMal-p2. Primer A and 5′-ATAGCTTCTAGATTAATTGTTAATTTCTCTGTCCC-3′(SEQ ID NO:15) (primer D) were used in the PCR with the template pgtat to generate the inserted DNA fragments. M41-P was used as the template with primer A and D in PCR to generate M41-PΔ178. All inserted sequences and mutated residues were checked by restriction enzyme analysis and confirmed by DNA sequencing.
  • 6.4.1.2 Purification and Characterization of Fusion Proteins
  • The fusion proteins were purified according to the protocol described in the manufacturer's brochure of protein fusion and purification systems from New England Biolabs (NEB). Fusion proteins (10 ng) were analyzed by electrophoresis on 8% SDS polyacrylamide gels. Western blotting analysis was performed as described by Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual, 2d Ed, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., Ch. 18, pp. 64-75. An HIV-1 positive serum diluted 1000-fold, or a human Fab derived from repertoire cloning was used to react with the fusion proteins. The second antibody was HRP-conjugated goat antihuman Fab. An ECL Western blotting detection system (Amersham) was used to detect the bound antibody. A detailed protocol for this detection system was provided by the manufacturer. Rainbow molecular weight markers (Amersham) were used to estimate the size of fusion proteins.
  • 6.4.1.3 Cell Fusion Assays for Anti-HIV Activity
  • Cell fusion assays were performed as previously described (Matthews et al., 1987, Proc. Natl. Acad. Sci. USA 84: 5424-5481). CEM cells (7×104) were incubated with HIV-1IIIB chronically infected CEM cells (104) in 96-well flat-bottomed half-area plates (Costar) in 100 culture medium. Peptide and fusion proteins at various concentrations in 10 μl culture medium were incubated with the cell mixtures at 37° C. for 24 hours. Multinucleated syncytia were estimated with microscopic examination. Both M41 and M41-P did not show cytotoxicity at the concentrations tested and shown in FIG. 8.
  • Inhibition of HIV-1 induced cell-cell fusion activity was carried out in the presence of 10 nM DP178 and various concentrations of M41Δ178 or M41-PΔ178 as indicated in FIG. 9. There was no observable syncytia in the presence of 10 nM DP178. No peptide or fusion protein was added in the control samples.
  • 6.4.1.4 ELISA Analysis of DP178 Binding to the Leucine Zipper Motif of GP41
  • The amino acid sequence of DP178 used is: YTSLIHSLIEESQNQQEKNEQELLELDKWASLWNWF(SEQ ID NO:1). For enzyme linked immunoassay (ELISA), M41Δ178 or M41-PΔ178 (5 μg/ml) in 0.1M NaHCO3, pH 8.6, were coated on 96 wells Linbro ELISA plates (Flow Lab, Inc.) overnight. Each well was washed three times with distilled water then blocked with 3% bovine serum albumin (BSA) for 2 hours. After blocking, peptides with 0.5% BSA in TBST (40 mM Tris-HCl pH7.5, 150 mM NaCl, 0.05% Tween 20) were added to the ELISA plates and incubated at room temperature for 1 hour. After washing three times with TBST, Fab-d was added at a concentration of 10 ng/ml with 0.5% BSA in TBST. The plates were washed three times with TBST after incubation at room temperature for 1 hour. Horse radish peroxidase (HRP) conjugated goat antihuman Fab antiserum at a 2000 fold dilution in TBST with 0.5% BSA was added to each well and incubated at room temperature for 45 minutes. The plates were then washed four times with TBST. The peroxidase substrate o-phenylene diamine (2.5 mg/ml) and 0.15% H2O2 were added to develop the color. The reaction was stopped with an equal volume of 4.5 NH2SO4 after incubation at room temperature for 10 minutes. The optical density of the stopped reaction mixture was measured with a micro plate reader (Molecular Design) at 490 nm. Results are shown in FIG. 10.
  • 6.4.2 Results 6.4.2.1 The Expression and Characterization of the Ectodomain of GP41
  • As a step toward understanding the roles of the two helical regions in gp41 structure and function, the ectodomain of gp41 was expressed as a maltose binding fusion protein (M41) (FIG. 7). The fusogenic peptide sequence at the N-terminal of gp41 was omitted from this recombinant protein and its derivatives to improve solubility. The maltose binding protein facilitated purification of the fusion proteins under relatively mild, non-denaturing conditions. Because the M41 soluble recombinant gp41 was not glycosylated, lacked several regions of the transmembrane protein (i.e., the fusion peptide, the membrane spanning, and the cytoplasmic domains), and was expressed in the absence of gp120, it was not expected to precisely reflect the structure of native gp41 on HIV-1 virions. Nevertheless, purified M41 folded in a manner that preserved certain discontinuous epitopes as evidenced by reactivity with human monoclonal antibodies, 98-6, 126-6, and 50-69, previously shown to bind conformational epitopes on native gp41 expressed in eukaryotic cells (Xu et al., 1991, J. Virol. 65: 4832-4838; Chen, 1994, J. Virol. 68:2002-2010). Thus, at least certain regions of native gp41 defined by these antibodies appear to be reproduced in the recombinant fusion protein M41. Furthermore, M41 reacted with a human recombinant Fab (Fab-d) that recognizes a conformational epitope on gp41 and binds HIV-1 virions as well as HIV-1 infected cells but not uninfected cells as analyzed by FACS. Deletion of either helix motif, i.e., DP107 or DP178, of the M41 fusion protein eliminated reactivity with Fab-d. These results indicate that both helical regions, separated by 60 amino acids in the primary sequence, are required to maintain the Fab-d epitope.
  • 6.4.2.2 Anti-HIV Activity of the Recombinant Ectodomain of GP41
  • The wild type M41 fusion protein was tested for anti-HIV-1 activity. As explained, supra, synthetic peptides corresponding to the leucine zipper (DP107) and the C-terminal putative helix (DP178) show potent anti-HIV activity. Despite inclusion of both these regions, the recombinant M41 protein did not affect HIV-1 induced membrane fusion at concentrations as high as 50 μM (Table XXV, below).
  • TABLE XXV
    DISRUPTION OF THE LEUCINE ZIPPER OF
    GP41 FREES THE ANTI-HIV MOTIF
    DP107 DP178 M41 M41-P M41-PΔ178
    Cell fusion
    1 μM  1 nM >50 μM 83 nM >50 μM
    (IC90)
    Fab-D 3.5 × 109 2.5 × 108
    binding (kD)
    HIV 1 μM 80 nM >16 μM 66 nM  >8 μM
    infectivity
    (IC90)
    1 The affinity constants of Fab-d binding to the fusion proteins were determined using a protocol described by B. Friguet et al., 1985, J. Immunol. Method. 77: 305-319.
    — = No detectable binding of Fab-d to the fusion proteins.
    Antiviral Infectivity Assays. 20 μl of serially diluted virus stock was incubated for 60 minutes at ambient temperature with 20 μl of the indicated concentration of purified recombinant fusion protein in RPMI 1640 containing 10% fetal bovine serum and antibiotics in a 96-well microtiter plate. 20 μl of CEM4 cells at 6 × 105 cells/ml were added to each well, and cultures were incubated at 37° C. in a humidified CO2 incubator. Cells were cultured for 9 days by the addition of fresh medium every 2 to 3 days. On days 5, 7, and 9 postinfection, supernatant samples were assayed for reverse transcriptase (RT) activity, as described below, to monitor viral replication. The 50% tissue culture infectious dose (TCID50) was calculated for each condition according to the formula of Reed & Muench, 1937, Am. J. Hyg. 27: 493-497. RT activity was determined by a modification of the published methods of Goff et al., 1981, J. Virol. 38: 239-248 and Willey et al., 1988, J. Virol. 62: 139-147 as described in Chen et al., 1993, AIDS Res. Human Retroviruses 9: 1079-1086.
  • Surprisingly, a single amino acid substitution, proline in place of isoleucine in the middle of the leucine zipper motif, yielded a fusion protein (M41-P) which did exhibit antiviral activity (Table XXV and FIG. 8). As seen in Table XXV, M41-P blocked syncytia formation by 90% at approximately 85 nM and neutralized HIV-1IIIB infection by 90% at approximately 70 nM concentrations. The anti-HIV-1 activity of M41-P appeared to be mediated by the C-terminal helical sequence since deletion of that region from M41-P yielded an inactive fusion protein, M41-PΔ178 (Table XXV). This interpretation was reinforced by experiments demonstrating that a truncated fusion protein lacking the DP178 sequence, M41Δ178, abrogated the potent anti-fusion activity of the DP178 peptide in a concentration-dependent manner (FIG. 9). The same truncated fusion protein containing the proline mutation disrupting the leucine zipper, M41-PΔ178, was not active in similar competition experiments (FIG. 9). The results indicate that the DP178 peptide associates with a second site on gp41 whose interactive structure is dependent on a wild type leucine zipper sequence. A similar interaction may occur within the wild type fusion protein, M41, and act to form an intramolecular clasp which sequesters the DP178 region, making it unavailable for anti-viral activity.
  • A specific association between these two domains is also indicated by other human monoclonal Fab-d studies. For example, Fab-d failed to bind either the DP178 peptide or the fusion protein M41Δ178, but its epitope was reconstituted by simply mixing these two reagents together (FIG. 10). Again, the proline mutation in the leucine zipper domain of the fusion protein, M41-PΔ178, failed to reconstitute the epitope in similar mixing experiments.
  • 6.5. Example: Method for Computer-Assisted Identification of DP107-Like and DP178-Like Sequences
  • A number of known coiled-coil sequences have been well described in the literature and contain heptad repeat positioning for each amino acid. Coiled-coil nomenclature labels each of seven amino acids of a heptad repeat A through G, with amino acids A and D tending to be hydrophobic positions. Amino acids E and G tend to be charged. These four positions (A, D, E, and G) form the amphipathic backbone structure of a monomeric alpha-helix. The backbones of two or more amphipathic helices interact with each other to form di-, tri-, tetrameric, etc., coiled-coil structures. In order to begin to design computer search motifs, a series of well characterized coiled coils were chosen including yeast transcription factor GCN4, Influenza Virus hemagglutinin loop 36, and human proto-oncogenes c-Myc, c-Fos, and c-Jun. For each peptide sequence, a strict homology for the A and D positions, and a list of the amino acids which could be excluded for the B, C, E, F, and G positions (because they are not observed in these positions) was determined. Motifs were tailored to the DP107 and DP178 sequences by deducing the most likely possibilities for heptad positioning of the amino acids of HIV-1 Bru DP-107, which is known to have coiled-coil structure, and HIV-1 Bru DP178, which is still structurally undefined. The analysis of each of the sequences is contained in FIG. 12. For example, the motif for GCN4 was designed as follows:
    • 1. The only amino acids (using standard single letter amino acid codes) found in the A or D positions of GCN4 were [LMNV].
    • 2. All amino acids were found at B, C, E, F, and G positions except {CFGIMPTW}.
    • 3. The PESEARCH motif would, therefore, be written as follows:
      • [LMNV]-{CFGIMPTW}(2)-[LMNV]-{CFGIMPTW}(3)-
      • [LMNV]-{CFGIMPTW}(2)-[LMNV]-{CFGIMPTW}(3)-
      • [LMNV]-{CFGIMPTW}(2)-[LMNV]-{CFGIMPTW}(3)-
      • [LMNV]-{CFGIMPTW}(2)-[LMNV]-{CFGIMPTW}(3)
        Translating or reading the motif: “at the first A position either L, M, N, or V must occur; at positions B and C (the next two positions) accept everything except C, F, G, I, M, P, T, or W; at the D position either L, M, N, or V must occur; at positions E, F, and G (the next 3 positions) accept everything except C, F, G, I, M, P, T, or W.” This statement is contained four times in a 28-mer motif and five times in a 35-mer motif. The basic motif key then would be: [LMNV]-{CFGIMPTW}. The motif keys for the remaining well described coiled-coil sequences are summarized in FIG. 12.
  • The motif design for DP107 and DP178 was slightly different than the 28-mer model sequences described above due to the fact that heptad repeat positions are not defined and the peptides are both longer than 28 residues. FIG. 13 illustrates several possible sequence alignments for both DP107 and DP178 and also includes motif designs based on 28-mer, 35-mer, and full-length peptides. Notice that only slight differences occur in the motifs as the peptides are lengthened. Generally, lengthening the base peptide results in a less stringent motif. This is very useful in broadening the possibilities for identifying DP107- or DP-178-like primary amino acid sequences referred to in this document as “hits”.
  • In addition to making highly specific motifs for each type peptide sequence to be searched, it is also possible to make “hybrid” motifs. These motifs are made by “crossing” two or more very stringent motifs to make a new search algorithm which will find not only both “parent” motif sequences but also any peptide sequences which have similarities to one, the other, or both “parents”. For example, in FIG. 14 the “parent” sequence of GCN4 is crossed with each of the possible “parent” motifs of DP-107. Now the hybrid motif must contain all of the amino acids found in the A and D positions of both parents, and exclude all of the amino acids not found in either parent at the other positions. The resulting hybrid from crossing GCN4 or [LMNV]{CFGIMPTW} and DP107 (28-mer with the first L in the D position) or [ILQT]{CDFIMPST}, is [ILMNQTV]{CFIMPT}. Notice that now only two basic hybrid motifs exist which cover both framing possibilities, as well as all peptide lengths of the parent DP-107 molecule. FIG. 15 represents the “hybridizations” of GCN4 with DP-178. FIG. 16 represents the “hybridizations” of DP107 and DP178. It is important to keep in mind that the represented motifs, both parent and hybrid, are motif keys and not the depiction of the full-length motif needed to actually do the computer search.
  • Hybridizations can be performed on any combination of two or more motifs. FIG. 17 summarizes several three-motif hybridizations including GCN4, DP107 (both frames), and DP178 (also both frames). Notice that the resulting motifs are now becoming much more similar to each other. In fact, the first and third hybrid motifs are actually subsets of the second and fourth hybrid motifs respectively. This means that the first and third hybrid motifs are slightly more stringent than the second and fourth. It should also be noted that with only minor changes in these four motifs, or by hybridizing them, a single motif could be obtained which would find all of the sequences. However, it should be remembered that stringency is also reduced. Finally, the most broad-spectrum and least-stringent hybrid motif is described in FIG. 18 which summarizes the hybridization of GCN4, DP107 (both frames), DP178 (both frames), c-Fos, c-Jun, c-Myc, and Flu loop 36.
  • A special set of motifs was designed based on the fact that DP-178 is located only approximately ten amino acids upstream of the transmembrane spanning region of gp41 and just C-terminal to a proline which separates DP107 and DP178. It has been postulated that DP178 may be an amphipathic helix when membrane associated, and that the proline might aid in the initiation of the helix formation. The same arrangement was observed in Respiratory Syncytial Virus; however, the DP178-like region in this virus also had a leucine zipper just C-terminal to the proline. Therefore, N-terminal proline-leucine zipper motifs were designed to analyze whether any other viruses might contain this same pattern. The motifs are summarized in FIG. 19.
  • The PC/Gene protein database contains 5879 viral amino acid sequences (library file PVIRUSES; CD-ROM release 11.0). Of these, 1092 are viral enveloped or glycoprotein sequences (library file PVIRUSE1). Tables V through XIV contain lists of protein sequence names and motif hit locations for all the motifs searched.
  • 6.6. Example: Computer-Assisted Identification of DP107 and DP178-Like Sequences in Human Immunodeficiency Virus
  • FIG. 20 represents search results for HIV-1 BRU isolate gp41 (PC/Gene protein sequence PENV_HV1BR). Notice that the hybrid motif which crosses DP-107 and DP-178 (named 107×178×4; the same motif as found in FIG. 16 found three hits including amino acids 550-599, 636-688, and 796-823. These areas include DP-107 plus eight N-terminal and four C-terminal amino acids; DP178 plus seven N-terminal and ten C-terminal amino acids; and an area inside the transmembrane region (cytoplasmic). FIG. 20 also contains the results obtained from searching with the motif named ALLMOTI5, for which the key is found in FIG. 17 ({CDGHP} {CFP}×5). This motif also found three hits including DP107 (amino acids 510-599), DP178 (615-717), and a cytoplasmic region (772-841). These hits overlap the hits found by the motif 107×178×4 with considerable additional sequences on both the amino and carboxy termini. This is not surprising in that 107×178×4 is a subset of the ALLMOTI5 hybrid motif. Importantly, even though the stringency of ALLMOTI5 is considerably less than 107×178×4, it still selectively identifies the DP107 and DP178 regions of gp41 shown to contain sequences for inhibitory peptides of HIV-1. The results of these two motif searches are summarized in Table V under the PC/Gene protein sequence name PENV_HV1BR. The proline-leucine zipper motifs also gave several hits in HIV-1 BRU including 503-525 which is at the very C-terminus of gp120, just upstream of the cleavage site (P7LZIPC and P12LZIPC); and 735-768 in the cytoplasmic domain of gp41 (P23LZIPC). These results are found in Tables VIII, IX, and X under the same sequence name as mentioned above. Notice that the only area of HIV-1 BRU which is predicted by the Lupas algorithm to contain a coiled-coil region, is from amino acids 635-670. This begins eight amino acids N-terminal to the start and ends eight amino acids N-terminal to the end of DP178. DP107, despite the fact that it is a known coiled coil, is not predicted to contain a coiled-coil region using the Lupas method.
  • 6.7. Example: Computer-Assisted Identification F DP107-Like and DP178-Like Sequences in Human Respiratory Syncytial Virus
  • FIG. 21 represents search results for Human Respiratory Syncytial Virus (RSV; Strain A2) fusion glycoprotein F1 (PC/Gene protein sequence name PVGLF_HRSVA). Motif 107×178×4 finds three hits including amino acids 152-202, 213-243, and 488-515. The arrangement of these hits is similar to what is found in HIV-1 except that the motif finds two regions with similarities to DP-178, one just downstream of what would be called the DP107 region or amino acids 213-243, and one just upstream of the transmembrane region (also similar to DP178) or amino acids 488-515. Motif ALLMOTI5 also finds three areas including amino acids 116-202, 267-302, and 506-549. The proline-leucine zipper motifs also gave several hits including amino acids 205-221 and 265-287 (P1LZIPC 265-280, P12LZIPC), and 484-513 (P7LZIPC and P12LZIPC 484-506, P23LZIPC). Notice that the PLZIP motifs also identify regions which share location similarities with DP-178 of HIV-1.
  • 6.8. Example: Computer-Assisted Identification of DP107-like and DP178-Like Sequences in Simian Immunodeficiency Virus
  • Motif hits for Simian immunodeficiency Virus gp41 (AGM3 isolate; PC/Gene protein sequence name PENV_SIVAG) are shown in FIG. 22. Motif 107×178×4 finds three hits including amino acids 566-593, 597-624, and 703-730. The first two hits only have three amino acids between them and could probably be combined into one hit from 566-624 which would represent a DP107-like hit. Amino acids 703 to 730 would then represent a DP178-like hit. ALLMOTI5 also finds three hits including amino acids 556-628 (DP107-like), 651-699 (DP178-like), and 808-852 which represents the transmembrane spanning region. SIV also has one region from 655-692 with a high propensity to form a coiled coil as predicted by the Lupas algorithm. Both 107×178×4 and ALLMOTI5 motifs find the same region. SIV does not have any PLZIP motif hits in gp41.
  • The identification of DP178/DP107 analogs for a second SIV isolate (MM251) is demonstrated in the Example presented, below, in Section 19.
  • 6.9. Example: Computer-Assisted Identification of DP107-Like and DP178 Like Sequences in Canine Distemper Virus
  • Canine Distemper Virus (strain Onderstepoort) fusion glycoprotein F1 (PC/Gene Protein sequence name PVGLF_CDVO) has regions similar to Human RSV which are predicted to be DP107-like and DP178-like (FIG. 23). Motif 107×178×4 highlights one area just C-terminal to the fusion peptide at amino acids 252-293. Amino acids 252-286 are also predicted to be coiled coil using the Lupas algorithm. Almost 100 amino acids C-terminal to the first region is a DP178-like area at residues 340-367. ALLMOTI5 highlights three areas of interest including: amino acids 228-297, which completely overlaps both the Lupas prediction and the DP107-like 107×178×4 hit; residues 340-381, which overlaps the second 107×178×4 hit; and amino acids 568-602, which is DP178-like in that it is located just N-terminal to the transmembrane region. It also overlaps another region (residues 570-602) predicted by the Lupas method to have a high propensity to form a coiled coil. Several PLZIP motifs successfully identified areas of interest including P6 and P12LZIPC which highlight residues 336-357 and 336-361 respectively; P1 and P12LZIPC which find residues 398-414; and P12 and P23LZIPC which find residues 562-589 and 562-592 respectively.
  • 6.10. Example: Computer-Assisted Identification of DP107-Like and DP178-Like Sequences in Newcastle Disease Virus
  • FIG. 24 shows the motif hits found in Newcastle Disease Virus (strain Australia-Victoria/32; PC Gene protein sequence name PVGLF_NDVA). Motif 107×178×4 finds two areas including a DP107-like hit at amino acids 151-178 and a DP178-like hit at residues 426-512. ALLMOTI5 finds three areas including residues 117-182, 231-272, and 426-512. The hits from 426-512 include a region which is predicted by the Lupas method to have a high coiled-coil propensity (460-503). The PLZIP motifs identify only one region of interest at amino acids 273-289 (P1 and 12LZIPC).
  • 6.11. Example: Computer-Assisted Identification of DP107-Like and DP178-Like Sequences in Human Parainfluenza Virus
  • Both motifs 107×178×4 and ALLMOTI5 exhibit DP107-like hits in the same region, 115-182 and 117-182 respectively, of Human Parainfluenza Virus (strain NIH 47885; PC/Gene protein sequence name PVGLF_p13H4; (FIG. 25). In addition, the two motifs have a DP178-like hit just slightly C-terminal at amino acids 207-241. Both motifs also have DP178-like hits nearer the transmembrane region including amino acids 457-497 and 462-512 respectively. Several PLZIP motif hits are also observed including 283-303 (P5LZIPC), 283-310 (P12LZIPC), 453-474 (P6LZIPC), and 453-481 (P23LZIPC). The Lupas algorithm predicts that amino acids 122-176 may have a propensity to form a coiled-coil.
  • 6.12. Example: Computer-Assisted Identification of DP107-Like and DP178-Like Sequences of Influenza A Virus
  • FIG. 26 illustrates the Lupas prediction for a coiled coil in Influenza A Virus (strain A/Aichi/2/68) at residues 379-436, as well as the motif hits for 107×178×4 at amino acids 387-453, and for ALLMOTI5 at residues 380-456. Residues 383-471 (38-125 of HA2) were shown by Carr and Kim to be an extended coiled coil when under acidic pH (Carr and Kim, 1993, Cell 73: 823-832). The Lupas algorithm predicts a coiled-coil at residues 379-436. All three methods successfully predicted the region shown to actually have coiled-coil structure; however, ALLMOTI5 predicted the greatest portion of the 88 residue stretch.
  • 6.13. Example: Potential Respiratory Syncytial Virus DP178/DP107 Analogs: CD and Antiviral Characterization
  • In the Example presented herein, respiratory syncytial virus (RSV) peptides identified by utilizing the computer-assisted search motifs described in the Examples presented in Sections 9 and 11, above, were tested for anti-RSV activity. Additionally, circular dichroism (CD) structural analyses were conducted on the peptides, as discussed below. It is demonstrated that several of the identified peptides exhibit potent antiviral capability. Additionally, it is shown that several of these peptides exhibit a substantial helical character.
  • 6.13.1 Materials and Methods Structural Analyses:
  • The CD spectra were measured in a 10 mM sodium phosphate, 150 mM sodium chloride, pH 7.0, buffer at approximately 10 mM concentrations, using a 1 cm pathlength cell on a Jobin/Yvon Autodichrograph Mark V CD spectrophotometer. Peptides were synthesized according to the methods described, above, in Section 6.1. Peptide concentrations were determined from A280 using Edlehoch's method (1967, Biochemistry 6:1948).
  • Anti-RSV antiviral activity assays: The assay utilized herein tested the ability of the peptides to disrupt the ability of HEp2 cells acutely infected with RSV (i.e., cells which are infected with a multiplicity of infection of greater than 2) to fuse and cause syncytial formation on a monolayer of uninfected an uninfected line of Hep-2 cells. The lower the observed level of fusion, the greater the antiviral activity of the peptide was determined to be.
  • Uninfected confluent monolayers of Hep-2 cells were grown in microtiter wells in 3% EMEM (Eagle Minimum Essential Medium w/o L-glutamine [Bio Whittaker Cat. No. 12-125F], with fetal bovine serum [FBS; which had been heat inactivated for 30 minutes at 56° C.; Bio Whittaker Cat. No. 14-501F) supplemented at 3%, antibiotics (penicillin/streptomycin; Bio Whittaker Cat. No. 17-602E) added at 1%, and glutamine added at 1%.
  • To prepare Hep2 cells for addition to uninfected cells, cultures of acutely infected Hep2 cells were washed with DPBS (Dulbecco's Phosphate Buffered Saline w/o calcium or magnesium; Bio Whittaker Cat. No. 17-512F) and cell monolayers were removed with Versene (1:5000; Gibco Life Technologies Cat. No. 15040-017). The cells were spun 10 minutes and resuspended in 3% FBS. Cell counts were performed using a hemacytometer. Persistent cells were added to the uninfected Hep-2 cells.
  • The antiviral assay was conducted by, first, removing all media from the wells containing uninfected Hep-2 cells, then adding peptides (at the dilutions described below) in 3% EMEM, and 100 acutely RSV-infected Hep2 cells per well. Wells were then incubated at 37° C. for 48 hours.
  • After incubation, cells in control wells were checked for fusion centers, media was removed from the wells, followed by addition, to each well, of either Crystal Violet stain or XTT. With respect to Crystal Violet, approximately 50 μl 0.25% Crystal Violet stain in methanol were added to each well. The wells were rinsed immediately, to remove excess stain, and were allowed to dry. The number of syncytia per well were then counted, using a dissecting microscope.
  • With respect to XTT (2,3-bis[2-Methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxyanilide inner salt), 50 μl XTT (1 mg/ml in RPMI buffered with 100 mM HEPES, pH 7.2-7.4, plus 5% DMSO) were added to each well. The OD450/690 was measured (after blanking against growth medium without cells or reagents, and against reagents) according to standard procedures.
  • Peptides: The peptides characterized in the study presented herein were:
  • 1) peptides T-142 to T-155 and T-575, as shown in FIG. 27A-C, and peptides T-22 to T-27, T-68, T-334 and T-371 to T-375 and T-575, as shown in FIG. 27C;
    2) peptides T-120 to T-141 and T-576, as shown in FIG. 27D-F, and peptides 1-12, T-13, T-15, T-19, T-28 to T-30, T-66, T-69, T-70 and T-576, as shown in FIG. 27F; and
    3) peptides T-67 and T-104 to T-119 and T-384, as shown in FIG. 28A-C, and peptides T-71, T-613 to T-617, T-662 to T-676 and T-730, as shown in FIG. 28C.
  • The peptides of group 1 represent portions of the RSV F2 protein DP178/107-like region. The peptides of group 2 represent portions of the RSV F1 protein DP107-like region. The peptides of groups 3 represent portions of the RSV F1 protein DP178-like region.
  • Each peptide was tested at 2-fold serial dilutions ranging from 100 μg/ml to approximately 100 ng/ml. For each of the assays, a well containing no peptide was also used. The IC50 data for each peptide represents the average of several experiments conducted utilizing that peptide.
  • 6.13.2 Results
  • The data summarized in FIGS. 27A-C and 28A-C represent antiviral and structural information obtained from peptides derived from the RSV F2 DP178/DP107-like F2 region (FIG. 27A-C), the RSV F1 DP-107-like region (FIG. 27D-F) and the RSV DP178-like F2 region (FIG. 28A-C).
  • As shown in FIGS. 27A-F, a number of the RSV DP178/DP107-like peptides exhibited a detectable level of antiviral activity. Peptides from the RSV DP178/DP107-like F2 region (FIG. 27A-C), for example, T-142 to T-145 and T-334 purfied peptides, exhibited detectable levels of antiviral activity, as evidenced by their IC50 values. Further, a number of RSV F1 DP107-like peptides (FIG. 27D-F) exhibited a sizable level of antiviral activity as purified peptides, including, for example, peptides T-124 to T-127, T-131, T-135 and T-137 to T-139, as demonstrated by their low IC50 values. In addition, CD analysis FIG. 27A-B, 27D-E) reveals that many of the peptides exhibit some detectable level of helical structure.
  • The results summarized in FIG. 28A-C demonstrate that a number of DP178-like purified peptides exhibit a range of potent anti-viral activity. These peptides include, for example, T-67, T-104, T-105 and T-107 to T-119, as listed in FIG. 28A-B, and T-665 to 1-669 and T-671 to 1-673, as listed in FIG. 28C. In addition, some of the DP178-like peptides exhibited some level of helicity.
  • Thus, the computer assisted searches described, hereinabove, successfully identified viral peptide domains that represent highly promising anti-RSV antiviral compounds.
  • 6.14. Example: Potential Human Parainfluenza Virus Type 3 DP178/DP107 Analogs: CD and Antiviral Characterization
  • In the Example presented herein, human parainfluenza virus type 3 (HPIV3) peptides identified by utilizing the computer-assisted search motifs described in the Examples presented in Sections 9 and 15, above, were tested for anti-HPIV3 activity. Additionally, circular dichroism (CD) structural analyses were conducted on the peptides, as discussed below. It is demonstrated that several of the identified peptides exhibit potent antiviral capability. Additionally, it is shown that several of these peptides exhibit a substantial helical character.
  • 6.14.1 Materials and Methods Structural Analyses:
  • Structural analyses consisted of circular dichroism (CD) studies. The CD spectra were measured in a 10 mM sodium phosphate, 150 mM sodium chloride, pH 7.0, buffer at approximately 10 mM concentrations, using a 1 cm pathlength cell on a Jobin/Yvon Autodichrograph Mark V CD spectrophotometer. Peptide concentrations were determined from A280 using Edlehoch's method (1967, Biochemistry 6:1948).
  • Anti-HPIV3 antiviral activity assays: The assay utilized herein tested the ability of the peptides to disrupt the ability of Hep2 cells chronically infected with HPIV3 to fuse and cause syncytial formation on a monolayer of an uninfected line of CV-1W cells. The more potent the lower the observed level of fusion, the greater the antiviral activity of the peptide.
  • Uninfected confluent monolayers of CV-1W cells were grown in microtiter wells in 3% EMEM (Eagle Minimum Essential Medium w/o L-glutamine [Bio Whittaker Cat. No. 12-125F], with fetal bovine serum [FBS; which had been heat inactivated for 30 minutes at 56° C.; Bio Whittaker Cat. No. 14-501F) supplemented at 3%, antibiotics/antimycotics (Gibco BRL Life Technologies Cat. No. 15040-017) added at 1%, and glutamine added at 1%.
  • To prepare Hep2 cells for addition to uninfected cells, cultures of chronically infected Hep2 cells were washed with DPBS (Dulbecco's Phosphate Buffered Saline w/o calcium or magnesium; Bio Whittaker Cat. No. 17-512F) and cell monolayers were removed with Versene (1:5000; Gibco Life Technologies Cat. No. 15040-017). The cells were spun 10 minutes and resuspended in 3% FBS. Cell counts were performed using a hemacytometer. Persistent cells were added to the uninfected CV-1W cells.
  • The antiviral assay was conducted by, first, removing all media from the wells containing uninfected CV-1W cells, then adding peptides (at the dilutions described below) in 3% EMEM, and 500 chronically HPIV3-infected Hep2 cells per well. Wells were then incubated at 37° C. for 24 hours.
  • On day 2, after cells in control wells were checked for fusion centers, media was removed from the wells, followed by addition, to each well, of approximately 50 μl 0.25% Crystal Violet stain in methanol. Wells were rinsed immediately, to remove excess stain and were then allowed to dry. The number of syncytia per well were then counted, using a dissecting microscope.
  • Alternatively, instead of Crystal Violet analysis, cells were assayed with XTT, as described, avove, in Section 17.1.
  • Peptides: The peptides characterized in the study presented herein were:
    • 1. Peptides 157 to 188, as shown in FIG. 29A-C, and peptides T-38 to T-40, T-42 to T-46 and T-582, as shown in FIG. 29D-E. These peptides are derived from the DP107 region of the HPIV3 F1 fusion protein (represented by HPF3 107, as shown in FIG. 29A-C); and
    • 2. Peptides 189 to 210, as shown in FIG. 30A-B, and T-269, T-626, T-383 and T-577 to T-579, as shown in FIG. 30C. These peptides are primarily derived from the DP178 region of the HPIV3 F1 fusion protein (represented by HPF3 178, as shown in FIG. 30A-B). Peptide T-626 contains two mutated amino acid resides (represented by a shaded background). Additionally, peptide T-577 represents F1 amino acids 65-100, T-578 represents F1 amino acids 207-242 and T-579 represents F1 amino acids 273-309.
  • Each peptide was tested at 2-fold serial dilutions ranging from 500 μg/ml to approximately 500 ng/ml. For each of the assays, a well containing no peptide was also used.
  • 6.14.2 Results
  • The data summarized in FIGS. 29A-E and 30A-C represent antiviral and structural information obtained from peptides derived from the HPIV3 fusion protein DP107-like region (FIG. 29A-E) and the HPIV3 fusion protein DP178-like region (FIG. 30A-C).
  • As shown in FIG. 29A-E, a number of the HPIV3 DP107-like peptides exhibited potent levels of antiviral activity. These peptides include, for example, peptides T-40, T-172 to T-175, T-178, T-184 and T-185.
  • CD analysis reveals that a number of the peptides exhibit detectable to substantial level of helical structure.
  • The results summarized in FIG. 30A-C demonstrate that a number of the DP178-like peptides tested exhibit a range of anti-viral activity. These peptides include, for example, peptides 194 to 211, as evidenced by their low IC50 values. In fact, peptides 201 to 205 exhibit IC50 values in the nanogram/ml range. In addition, many of the DP178-like peptides exhibited some level of helicity.
  • Thus, the computer assisted searches described, hereinabove, have successfully identified viral peptide domains that represent highly promising anti-HPIV3 antiviral compounds.
  • 6.15. Example: Computer-Assisted Identification of DP178/DP107 Analogs in Simian Immunodeficiency Virus
  • FIG. 31 represents search results for SIV isolate MM251 (PC/Gene® protein sequence PENV_SIVM2). Both 107×178×4 and ALLMOTI5 search motifs identified two regions with similarities to DP107 and/or DP178.
  • The peptide regions found by 107×178×4 were located at amino acid residues 156-215 and 277-289. The peptide regions found by ALLMOTI5 were located at amino acid residues 156-219 and 245-286. Both motifs, therefore, identify similar regions.
  • Interestingly, the first SIV peptide region (i.e., from amino acid residue 156 to approximately amino acid residue 219) correlates with a DP107 region, while the second region identified (i.e., from approximately amino acid residue 245 to approximately amino acid residue 289) correlates with the DP178 region of HIV. In fact, an alignment of SIV isolate MM251 and HIV isolate BRU, followed by a selection of the best peptide matches for HIV DP107 and DP178, reveals that the best matches are found within the peptide regions identified by the 107×178×4 and ALLMOTI5 search motifs.
  • It should be noted that a potential coiled-coil region at amino acid residues 242-282 is predicted by the Lupas program. This is similar to the observation in HIV in which the coiled-coil is predicted by the Lupas program to be in the DP178 rather than in the DP107 region. It is possible, therefore, that SIV may be similar to HIV in that it may contain a coiled-coil structure in the DP107 region, despite such a structure being missed by the Lupas algorithm. Likewise, it may be that the region corresponding to a DP178 analog in SIV may exhibit an undefined structure, despite the Lupas program's prediction of a coiled-coil structure.
  • 6.16. Example: Computer-Assisted Identification of DP178/DP107 Analogs in Epstein-Barr Virus
  • The results presented herein describe the identification of DP178/DP107 analogs within two different Epstein-Barr Virus proteins. Epstein-Barr is a human herpes virus which is the causative agent of, for example, infectious mononucleosis (IM), and is also associated with nasopharyngeal carcinomas (NPC), Burkitt's lymphoma and other diseases. The virus predominantly exists in the latent form and is activated by a variety of stimuli.
  • FIG. 32 depicts the search motif results for the Epstein-Barr Virus (Strain B95-8; PC/Gene® protein sequence PVGLB_EBV) glycoprotein gp110 precursor (gp115). The 107×178×4 motif identified two regions of interest, namely the regions covered by amino acid residues 95-122 and 631-658. One PZIP region was identified at amino acid residue 732-752 which is most likely a cytoplasmic region of the protein. The Lupas algorithm predicts a coiled-coil structure for amino acids 657-684. No ALLMOTI5 regions were identified.
  • FIG. 33 depicts the search motif results for the Zebra (or EB1) trans-activator protein (BZLF1) of the above-identified Epstein-Barr virus. This protein is a transcription factor which represents the primary mediator of viral reactivation. It is a member of the b-ZIP family of transcription factors and shares significant homology with the basic DNA-binding and dimerization domains of the cellular oncogenes c-fos and C/EBP. The Zebra protein functions as a homodimer.
  • Search results domonstrate that the Zebra protein exhibits a single region which is predicted to be either of DP107 or DP178 similarity, and is found between the known DNA binding and dimerization regions of the protein. Specifically, this region is located at amino acid residues 193-220, as shown in FIG. 33. The Lupas program predicted no coiled-coil regions.
  • 6.17. Example: Computer-Assisted Identification of DP178/DP107 Analogs in Measles Virus
  • FIG. 34 illustrates the motif search results for the fusion protein F1 of measles virus, strain Edmonston (PC Gene® protein sequence PVGLF MEASE), successfully identifying DP178/DP107 analogs.
  • The 107×178×4 motif identifies a single region at amino acid residues 228-262. The ALLMOTI5 search motif identifies three regions, including amino acid residues 116-184, 228-269 and 452-500. Three regions containing proline residues followed by a leucine zipper-like sequence were found beginning at proline residues 214, 286 and 451.
  • The Lupas program identified two regions it predicted had potential for coiled-coil structure, which include amino acid residues 141-172 and 444-483.
  • 6.18. Example: Computer-Assisted Identification of DP178/DP107 Analogs in Hepatitis B Virus
  • FIG. 35 depicts the results of a PZIP motif search conducted on the Hepatitis B virus subtype AYW. Two regions of interest within the major surface antigen precursor S protein were identified. The first lies just C-terminal to the proposed fusion peptide of the major surface antigen (Hbs) which is found at amino acid residues 174-191. The second region is located at amino acid residues 233-267. The Lupas program predicts no coiled-coil repeat regions.
  • In order to test the potential anti-HBV antiviral activity of these D178/DP107 analog regions, peptides derived from area around the analog regions are synthesized, as shown in FIG. 52A-B. These peptides represent one amino acid peptide “walks” through the putative DP178/DP107 analog regions. The peptides are synthesized according to standard Fmoc chemistry on Rinkamide MBHA resins to provide for carboxy terminal blockade (Chang, C. D. and Meinhofer, J., 1978, Int. J. Pept. Protein Res. 11:246-249; Fields, G. B. and Noble, R. L., 1990, Int. J. Pept. Protein Res. 35:161-214). Follwing complete synthesis, the peptide amino-terminus is blocked through automated acetylation and the peptide is cleaved with trifluoroacetic acid (TFA) and the appropriate scavengers (King, D. S. et al., 1990, Int. J. Pept. Res. 36:255-266). After cleavage, the peptide is precipitated with ether and dried under vacuum for 24 hours.
  • The anti-HBV activity of the peptides is tested by utilizing standard assays to determine the test peptide concentration required to cause an acceptable (e.g., 90%) decrease in the amount of viral progeny formed by cells exposed to an HBV viral inoculum. Candidate antivial peptides are further characterized in model systems such as wood chuck tissue culture and animal sytems, prior to testing on humans.
  • 6.19. Example: Computer-Assisted Identification of DP178/DP107 Analogs in Simian Mason-Pfizer Monkey Virus
  • The results depicted herein illustrate the results of search motifs conducted on the Simian Mason-Pfizer monkey virus. The motifs reveal DP178/DP107 analogs within the enveloped (TM) protein GP20, as shown in FIG. 36.
  • The 107×178×4 motifs identifies a region at amino acid residues 422-470. The ALLMOTI5 finds a region at amino acid residues 408-474. The Lupas program predicted a coiled-coil structure a amino acids 424-459.
  • 6.20. Example: Computer-Assisted Identification of DP178/DP107 Analogs in Bacterial Proteins
  • The results presented herein demonstrate the identification of DP178/DP107 analogs corresponding to sequences present in proteins of a variety of bacterial species.
  • FIG. 37 depicts the search motif results for the Pseudomonas aeruginosa fimbrial protein (Pilin). Two regions were identified by motifs 107×178×4 and ALLMOTI5. The regions located at amino acid residues 30-67 and 80-144 were identified by the 107×178×4 motif. The regions at amino acid residues 30-68 and 80-125 were identified by the ALLMOTI5.
  • FIG. 38 depicts the search motif results for the Pseudomonas gonorrhoeae fimbrial protein (Pilin). A single region was identified by both the 107×178×4 and the ALLMOTI5 motifs. The region located at amino acid residues 66-97 was identified by the 107×178×4 motif. The region located at amino acid residues 66-125 were identified by the ALLMOTI5 search motif. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 39 depicts the search motif results for the Hemophilus Influenza fimbrial protein (Pilin). A single region was identified by both the 107×178×4 and the ALLMOTI5 motifs. The region located at amino acid residues 102-129 was identified by the 107×178×4 motif. The region located at amino acid residues 102-148 were identified by the ALLMOTI5 search motif. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 40 depicts the search motif results for the Staphylococcus aureus toxic shock syndrome Hemophilus Influenza fimbrial protein (Pilin). A single region was identified by both the 107×178×4 and the ALLMOTI5 motifs. The region located at amino acid residues 102-129 was identified by the 107×178×4 motif. The region located at amino acid residues 102-148 were identified by the ALLMOTI5 search motif. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 41 summarizes the motif search results conducted on the Staphylococcus aureus enterotoxin Type E protein. These results demonstrate the successful identification of DP178/DP107 analogs corresponding to peptide sequences within this protein, as described below.
  • The ALLMOTI5 motif identified a region at amino acid residues 22-27. The 107×178×4 motif identified two regions, with the first at amino acid residues 26-69 and the second at 88-115. A P12LZIPC motif search identified two regions, at amino acid residues 163-181 and 230-250.
  • The Lupas program predicted a region with a high propensity for coiling at amino acid residues 25-54. This sequence is completely contained within the first region identified by both ALLMOTI5 and 107×178×4 motifs.
  • FIG. 42 depicts the search motif results conducted on a second Staphylococcus aureus toxin, enterotoxin A. Two regions were identified by the ALLMOTI5 motif, at amino acid residues 22-70 and amino acid residues 164-205. The 107×178×4 motif found two regions, the first at amino acid residues 26-69 and the second at amino acid residues 165-192. A P23LZIPC motif search revealed a region at amino acid residues 216-250. No coiled-coil regions were predicted by the Lupas program.
  • FIG. 43 shows the motif search results conducted on the E. coli heat labile enterotoxin A protein, demonstrating that identification of DP178/DP107 analogs corresponding to peptides located within this protein. Two regions were identified by the ALLMOTI5 motif, with the first residing at amino acid residues 55-115, and the second residing at amino acid residues 216-254. The 107×178×4 motif identified a single region at amino acid residues 78-105. No coiled-coil regions were predicted by the Lupas program.
  • 6.21. Example: Computer-Assisted Identification of DP178/DP107 Analogs within Various Human Proteins
  • The results presented herein demonstrate the identification of DP178/DP107 analogs corresponding to peptide sequences present within several different human proteins.
  • FIG. 44 illustrates the search motif results conducted on the human c-fos oncoprotein. The ALLMOTI5 motif identified a single region at amino acid residues 155-193. The 107×178×4 motif identified one region at amino acid residues 162-193. The Lupas program predicted a region at amino acid residues 148-201 to have coiled-coil structure.
  • FIG. 45 illustrates the search motif results conducted on the human lupus KU autoantigen protein P70. The ALLMOTI5 motif identified a single region at amino acid residues 229-280. The 107×178×4 motif identified one region at amino acid residues 235-292. The Lupas program predicted a region at amino acid residues 232-267 to have coiled-coil structure.
  • FIG. 46 illustrates the search motif results conducted on the human zinc finger protein 10. The
  • ALLMOTI5 motif identified a single region at amino acid residues 29-81. The 107×178×4 motif identified one region at amino acid residues 29-56. A P23LZIPC motif search found a single region at amino acid residues 420-457. The Lupas program predicted no coiled-coil regions.
  • 6.22. Example: Potential Measles Virus DP178/DP107 Analogs: CD and Antiviral Characterization
  • In the Example presented herein, measles (MeV) virus DP178-like peptides identified by utilizing the computer-assisted search motifs described in the Examples presented in Sections 9 and 21, above, are tested for anti-MeV activity. Additionally, circular dichroism (CD) structural analyses are conducted on the peptides, as discussed below. It is demonstrated that several of the identified peptides exhibit potent antiviral capability. Additionally, it is shown that none of the these peptides exhibit a substantial helical character.
  • 6.22.1 Materials and Methods Structural Analyses:
  • The CD spectra were measured in a 10 mM sodium phosphate, 150 mM sodium chloride, pH 7.0, buffer at approximately 10 mM concentrations, using a 1 cm pathlength cell on a Jobin/Yvon Autodichrograph Mark V CD spectrophotometer. Peptide concentrations were determined from A280 using Edlehoch's method (1967, Biochemistry 6:1948).
  • Anti-MeV antiviral activity syncytial reduction assay: The assay utilized herein tested the ability of the peptides to disrupt the ability of Vero cells acutely infected with MeV (i.e., cells which are infected with a multiplicity of infection of 2-3) to fuse and cause syncytial formation on a monolayer of an uninfected line of Vero cells. The more potent the peptide, the lower the observed level of fusion, the greater the antiviral activity of the peptide.
  • Uninfected confluent monolayers of Vero cells were grown in microtiter wells in 10% FBS EMEM (Eagle Minimum Essential Medium w/o L-glutamine [Bio Whittaker Cat. No. 12-125F], with fetal bovine serum [FBS; which had been heat inactivated for 30 minutes at 56° C.; Bio Whittaker Cat. No. 14-501F) supplemented at 10%, antibiotics/antimycotics (Bio Whittaker Cat. No. 17-602E) added at 1%, and glutamine added at 1%.
  • To prepare acutely infected Vero cells for addition to the uninfected cells, cultures of acutely infected Vero cells were washed twice with HBSS (Bio Whittaker Cat. No. 10-543F) and cell monolayers were removed with trypsin (Bio Whittaker Cat. No. 17-161E). Once cells detached, media was added, any remaining clumps of cells were dispersed, and hemacytometer cell counts were performed.
  • The antiviral assay was conducted by, first, removing all media from the wells containing uninfected Vero cells, then adding peptides (at the dilutions described below) in 10% FBS EMEM, and 50-100 acutely MeV-infected Vero cells per well. Wells were then incubated at 37° C. for a maximum of 18 hours.
  • On day 2, after cells in control wells were checked for fusion centers, media was removed from the wells, followed by addition, to each well, of approximately 50 μl 0.25% Crystal Violet stain in methanol. Wells were rinsed twice with water immediately, to remove excess stain and were then allowed to dry. The number of syncytia per well were then counted, using a dissecting microscope.
  • Anti-MeV antiviral activity plaque reduction assay: The assay utilized herein tested the ability of the peptides to disrupt the ability of MeV to infect permissive, uninfected Vero cells, leading to the infected cells' fusing with uninfected cells to produce syncytia. The lower the observed level of syncytial formation, the greater the antiviral activity of the peptide.
  • Monolayers of uninfected Vero cells are grown as described above.
  • The antiviral assay was conducted by, first, removing all media from the wells containing uninfected Vero cells, then adding peptides (at the dilutions described below) in 10% FBS EMEM, and MeV stock virus at a final concentration of 30 plaque forming units (PFU) per well. Wells were then incubated at 37° C. for a minimum of 36 hours and a maximum of 48 hours.
  • On day 2, after cells in control wells were checked for fusion centers, media was removed from the wells, followed by addition, to each well, of approximately 50 μl 0.25% Crystal Violet stain in methanol. Wells were rinsed twice with water immediately, to remove excess stain and were then allowed to dry. The number of syncytia per well were then counted, using a dissecting microscope.
  • Peptides: The peptides characterized in the study presented herein were peptides T-252A0 to T-256A0, T-25781/C1, and T-258B1 to T-265B0, and T-266A0 to T-268A0, as shown in FIG. 47A-B. These peptides represent a walk through the DP178-like region of the MeV fusion protein.
  • Each peptide was tested at 2-fold serial dilutions ranging from 100 μg/ml to approximately 100 ng/ml. For each of the assays, a well containing no peptide was also used.
  • 6.22.2 Results
  • The data summarized in FIG. 47A-B represents antiviral and structural information obtained via “peptide walks” through the DP178-like region of the MeV fusion protein.
  • As shown in FIG. 47A-B, the MeV DP178-like peptides exhibited a range of antiviral activity as crude peptides. Several of these peptides were chosen for purification and further antiviral characterization. The IC50 values for such peptides were determined, as shown in FIG. 47A-B, and ranged from 1.35 μg/ml (T-257B1/C1) to 0.072 μg/ml (T-265B1). None of the DP178-like peptides showed, by CD analysis, a detectable level of helicity.
  • Thus, the computer assisted searches described, hereinabove, as in for example, the Example presented in Section 9, for example, successfully identified viral peptide domains that represent highly promising anti-MeV antiviral compounds.
  • 6.23. Example: Potential SIV DP178/DP107 Analogs: Antiviral Characterization
  • In the Example presented herein, simian immunodeficiency virus (SIV) DP178-like peptides identified by utilizing the computer-assisted search motifs described in the Examples presented in Sections 9, 12 and 19, above, were tested for anti-SIV activity. It is demonstrated that several of the identified peptides exhibit potent antiviral capability.
  • 6.23.1 Materials and Methods Anti-SIV Antiviral Assays:
  • The assay utilized herein were as reported in Langolis et al. (Langolis, A. J. et al., 1991, AIDS Research and Human Retroviruses 7:713-720).
  • Peptides: The peptides characterized in the study presented herein were peptides T-391 to T-400, as shown in FIG. 48A-B. These peptides represent a walk through the DP178-like region of the SIV TM protein.
  • Each peptide was tested at 2-fold serial dilutions ranging from 100 μg/ml to approximately 100 ng/ml. For each of the assays, a well containing no peptide was also used.
  • 6.23.2 Results
  • The data summarized in FIG. 48A-B represents antiviral information obtained via “peptide walks” through the DP178-like region of the SIV TM protein.
  • As shown in FIG. 48A-B, peptides T-391 to T-400 were tested and exhibited a potent antiviral activity as crude peptides.
  • Thus, the computer assisted searches described, hereinabove, as in for example, the Example presented in Section 9, for example, successfully identified viral peptide domains that represent highly promising anti-SIV antiviral compounds.
  • 6.24. Example: Anti-Viral Activity of DP107 and DP-178 Peptide Truncations and Mutations
  • The Example presented in this Section represents a study of the antiviral activity of DP107 and DP178 truncations and mutations. It is demonstrated that several of these DP107 and DP178 modified peptides exhibit substantial antiviral activity.
  • 6.24.1 Materials and Methods
  • Anti-HIV assays: The antiviral assays performed were as those described, above, in Section 6.1. Assays utilized HIV-1/IIIb and/or HIV-2 NIHZ isolates. Purified peptides were used, unless otherwise noted in FIGS. 49A-L.
  • Peptides: The peptides characterized in the study presented herein were:
    • 1. FIGS. 49A-L present peptides derived from the region around and containing the DP178 region of the HIV-1 BRU isolate. Specifically, this region spanned from gp41 amino acid residue 615 to amino acid residue 717. The peptides listed contain truncations of this region and/or mutations which vary from the DP178 sequence amino acid sequence. Further, certain of the peptides have had amino- and/or carboxy-terminal groups either added or removed, as indicated in the figures; and
    • 2. FIG. 50A-B. presents peptides which represent truncations of DP107 and/or the gp41 region surrounding the DP107 amino acid sequence of HIV-1 BRU isolate. Certain of the peptides are unblocked or biotinylated, as indicated in the figure.
      Blocked peptides contained an acyl N-terminus and an amido C-terminus.
    6.24.2 Results
  • Anti-HIV antiviral data was obtained with the group 1 DP178-derived peptides listed in FIG. 49A-L. The full-length, non-mutant DP178 peptide (referred to in FIG. 49A-L as T20) results shown are for 4 ng/ml.
  • In FIG. 49A-D, a number of the DP178 truncations exhibited a high level of antiviral activity, as evidenced by their low IC50 values. These include, for example, test peptides T-50, T-624, T-636 to T-641, T-645 to T-650, T-652 to T-654 and T-656. T-50 represents a test peptide which contains a point mutation, as indicated by the residue's shaded background. The HIV-1-derived test peptides exhibited a distinct strain-specific antiviral activity, in that none of the peptides tested on the HIV-2 NIHZ isolate demonstrated appreciable antti-HIV-2 antiviral activity.
  • Among the peptides listed in FIG. 49E-H, are test peptides representing the amino (T-4) and carboxy (T-3) terminal halves of DP178 were tested. The amino terminal peptide was not active (IC50>400 μg/ml) whereas the carboxy terminal peptide showed potent antiviral activity (IC50=3 μg/ml). A number of additional test peptides also exhibited a high level of antiviral activity. These included, for example, T-61/T-102, T-217 to T-221, T-235, T-381, T-677, T-377, T-590, T-378, T-591, T-271 to T-272, T-611, T-222 to T-223 and T-60/T-224. Certain of the antiviral peptides contain point mutations and/or amino acid residue additions which vary from the DP178 amino acid sequence.
  • In FIG. 49I-L, point mutations and/or amino and/or carboxy-terminal modifications are introduced into the DP178 amino acid sequence itself. As shown in the figure, the majority of the test peptides listed exhibit potent antiviral activity.
  • Truncations of the DP107 peptide (referred to in FIG. 50 as T21) were also produced and tested, as shown in FIG. 50A-B. FIG. 50A-B also presents data concerning blocked and unblocked peptides which contain additional amino acid residues from the gp41 region in which the DP107 sequence resides. Most of these peptides showed antiviral activity, as evidenced by their low IC50 values.
  • Thus, the results presented in this Section demonstrate that not only do the full length DP107 and DP178 peptides exhibit potent antiviral activity, but truncations and/or mutant versions of these peptides can also possess substantial antiviral character.
  • 6.25. Example: Potential Epstein-Barr DP178/DP107 Analogs: Antiviral Characterization
  • In the Example presented herein, peptides derived from the Epstein-Barr (EBV) DP-178/DP107 analog region of the Zebra protein identified, above, in the Example presented in Section 20 are described and tested for anti-EBV activity. It is demonstrated that among these peptides are ones which exhibit potential anti-viral activity.
  • 6.25.1 Materials and Methods
  • Electrophoretic Mobility Shift Assays (EMSA): Briefly, an EBV Zebra protein was synthesized utilizing SP6 RNA polymerase in vitro transcription and wheat germ in vitro translation systems (Promega Corporation recommendations; Butler, E. T. and Chamberlain, M. J., 1984, J. Biol. Chem. 257:5772; Pelham, H. R. B. and Jackson, R. J., 1976, Eur. J. Biochem. 67:247). The in vitro translated Zebra protein was then preincubated with increasing amounts of peptide up to 250 ng/ml prior to the addition of 10,000 to 20,000 c.p.m. of a 32P-labeled Zebra response element DNA fragment. After a 20 minute incubation in the presence of the response element, the reaction was analyzed on a 4% non-denaturing polyacrylamide gel, followed by autoradiography, utilizing standard gel-shift procedures. The ability of a test peptide to prevent Zebra homodimer DNA binding was assayed by the peptide's ability to abolish the response element gel migration retardation characteristic of a protein-bound nucleic acid molecule.
  • Peptides: The peptides characterized in this study represent peptide walks through the region containing, and flanked on both sides by, the DP178/DP107 analog region identified in the Example presented in Section 20, above, and shown as shown in FIG. 33. Specifically, the peptide walks covered the region from amino acid residue 173 to amino acid residue 246 of the EBV Zebra protein.
  • Each of the tested peptides were analyzed at a range of concentrations, with 150 ng/ml being the lowest concentration at which any of the peptides exerted an inhibitory effect.
  • 6.25.2 Results
  • The EBV Zebra protein transcription factor contains a DP178/DP107 analog region, as demonstrated in the Example presented, above, in Section 20. This protein appears to be the primary factor responsible for the reactivation capability of the virus. A method by which the DNA-binding function of the Zebra virus may be abolished may, therefore, represent an effective antiviral technique. In order to identify potential anti-EBV DP178/DP107 peptides, therefore, peptides derived from the region identified in Section 20, above, were tested for their ability to inhibit Zebra protein DNA binding.
  • The test peptides' ability to inhibit Zebra protein DNA binding was assayed via the EMSA assays described, above, in Section 28.1. The data summarized in FIG. 51A-C presents the results of EMSA assays of the listed EBV test peptides. These peptides represent one amino acid “walks” through the region containing, and flanked on both sides by, the DP178/DP107 analog region identified in the Example presented in Section 20, above, and shown as shown in FIG. 33. As shown in FIG. 51A-C, the region from which these peptides are derived lies from EBV Zebra protein amino acid residue 173 to 246. A number of the test peptides which were assayed exhibited an ability to inhibit Zebra protein homodimer DNA binding, including 439, 441, 444 and 445.
  • Those peptides which exhibit an ability to inhibit Zebra protein DNA binding represent potential anti-EBV antiviral compounds whose ability to inhibit EBV infection can be further characterized.
  • The present invention is not to be limited in scope by the specific embodiments described which are intended as single illustrations of individual aspects of the invention, and functionally equivalent methods and components are within the scope of the invention. Indeed, various modifications of the invention, in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.
  • LENGTHY TABLES
    The patent application contains a lengthy table section. A copy of the table is available in electronic form from the USPTO web site (http://seqdata.uspto.gov/?pageRequest=docDetail&DocID=US20100291680A1). An electronic copy of the table will also be available from the USPTO upon request and payment of the fee set forth in 37 CFR 1.19(b)(3).

Claims (15)

1. An isolated peptide recognized by an ALLMOTI5, 107×178×4 or a PLZIP sequence search motif
2. The peptide of claim 1 wherein the peptide corresponds to a peptide present in a virus.
3. The peptide of claim 2 in which the virus is HIV-1 or HIV-2.
4. The peptide of claim 2 in which the virus is a respiratory syncytial virus.
5. The peptide of claim 2 in which the virus is a human parainfluenza virus.
6. The peptide of claim 2 in which the virus is an influenza virus.
7. The peptide of claim 2 in which the virus is a hepatitis B virus.
8. The peptide of claim 2 wherein the virus is an Epstein-Barr virus.
9. A method for the inhibition of transmission of a virus to a cell, comprising contacting the cell with an effective concentration of a peptide recognized by an ALLMOTI5, 107×178×4 or a PLZIP sequence search motif for an effective period of time so that no infection of the cell by the virus occurs.
10. The method of claim 9 wherein the virus is HIV-1 or HIV-2.
11. The method of claim 9 wherein the virus is a respiratory syncytial virus.
12. The method of claim 9 wherein the virus is a human parainfluenza virus.
13. The method of claim 9 wherein the virus is an influenza virus.
14. The method of claim 9 in which the virus is a hepatitis B virus.
15. The method of claim 9 wherein the virus is an Epstein-Barr virus.
US12/791,983 1994-06-07 2010-06-02 Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission Abandoned US20100291680A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/791,983 US20100291680A1 (en) 1994-06-07 2010-06-02 Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/255,208 US6440656B1 (en) 1993-06-07 1994-06-07 Methods for the inhibition of respiratory syncytial virus transmission
US08/360,107 US6017536A (en) 1993-06-07 1994-12-20 Simian immunodeficiency virus peptides with antifusogenic and antiviral activities
US08/470,896 US6479055B1 (en) 1993-06-07 1995-06-06 Methods for inhibition of membrane fusion-associated events, including respiratory syncytial virus transmission
US08/484,223 US7794725B1 (en) 1993-06-07 1995-06-07 Isolated peptides derived from human immunodeficiency virus types 1 and 2 containing fusion inhibitory domains
US12/791,983 US20100291680A1 (en) 1994-06-07 2010-06-02 Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/484,223 Continuation US7794725B1 (en) 1993-06-07 1995-06-07 Isolated peptides derived from human immunodeficiency virus types 1 and 2 containing fusion inhibitory domains

Publications (1)

Publication Number Publication Date
US20100291680A1 true US20100291680A1 (en) 2010-11-18

Family

ID=27000760

Family Applications (17)

Application Number Title Priority Date Filing Date
US08/470,896 Expired - Lifetime US6479055B1 (en) 1993-06-07 1995-06-06 Methods for inhibition of membrane fusion-associated events, including respiratory syncytial virus transmission
US08/487,266 Expired - Lifetime US6824783B1 (en) 1993-06-07 1995-06-07 Methods for inhibition of membrane fusion-associated events, including HIV transmission
US08/475,668 Expired - Fee Related US6060065A (en) 1993-06-07 1995-06-07 Compositions for inhibition of membrane fusion-associated events, including influenza virus transmission
US08/484,741 Expired - Fee Related US6951717B1 (en) 1993-06-07 1995-06-07 Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission
US08/485,551 Expired - Fee Related US6068973A (en) 1993-06-07 1995-06-07 Methods for inhibition of membrane fusion-associated events, including influenza virus
US08/474,349 Expired - Fee Related US6333395B1 (en) 1993-06-07 1995-06-07 Compositions for inhibition of membrane fusion-associated events, including human parainfluenza virus transmission
US08/471,913 Expired - Fee Related US6093794A (en) 1993-06-07 1995-06-07 Isolated peptides derived from the Epstein-Barr virus containing fusion inhibitory domains
US08/486,099 Expired - Fee Related US6013263A (en) 1993-06-07 1995-06-07 Measles virus peptides with antifusogenic and antiviral activities
US08/484,223 Expired - Fee Related US7794725B1 (en) 1993-06-07 1995-06-07 Isolated peptides derived from human immunodeficiency virus types 1 and 2 containing fusion inhibitory domains
US08/485,264 Expired - Fee Related US6228983B1 (en) 1993-06-07 1995-06-07 Human respiratory syncytial virus peptides with antifusogenic and antiviral activities
US08/919,597 Expired - Fee Related US6054265A (en) 1993-06-07 1997-09-26 Screening assays for compounds that inhibit membrane fusion-associated events
US10/267,748 Expired - Fee Related US7122190B2 (en) 1993-06-07 2002-10-08 Fusion proteins comprising DP-178 and other viral fusion inhibitor peptides useful for treating aids
US10/267,682 Expired - Fee Related US7273614B2 (en) 1993-06-07 2003-01-06 Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids
US10/739,355 Abandoned US20070037141A1 (en) 1994-06-07 2003-12-17 Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission
US11/253,274 Expired - Fee Related US7988974B2 (en) 1993-06-07 2005-10-17 Antifusogenic proteins comprising human immunodeficiency virus type 1 (HIV-1) gp41 DP-178 polypeptide variants and a macromolecular carrier
US12/791,983 Abandoned US20100291680A1 (en) 1994-06-07 2010-06-02 Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission
US13/161,919 Abandoned US20110275146A1 (en) 1993-06-07 2011-06-16 Fusion proteins comprising dp-178 and other viral fusion inhibitor peptides useful for treating aids

Family Applications Before (15)

Application Number Title Priority Date Filing Date
US08/470,896 Expired - Lifetime US6479055B1 (en) 1993-06-07 1995-06-06 Methods for inhibition of membrane fusion-associated events, including respiratory syncytial virus transmission
US08/487,266 Expired - Lifetime US6824783B1 (en) 1993-06-07 1995-06-07 Methods for inhibition of membrane fusion-associated events, including HIV transmission
US08/475,668 Expired - Fee Related US6060065A (en) 1993-06-07 1995-06-07 Compositions for inhibition of membrane fusion-associated events, including influenza virus transmission
US08/484,741 Expired - Fee Related US6951717B1 (en) 1993-06-07 1995-06-07 Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission
US08/485,551 Expired - Fee Related US6068973A (en) 1993-06-07 1995-06-07 Methods for inhibition of membrane fusion-associated events, including influenza virus
US08/474,349 Expired - Fee Related US6333395B1 (en) 1993-06-07 1995-06-07 Compositions for inhibition of membrane fusion-associated events, including human parainfluenza virus transmission
US08/471,913 Expired - Fee Related US6093794A (en) 1993-06-07 1995-06-07 Isolated peptides derived from the Epstein-Barr virus containing fusion inhibitory domains
US08/486,099 Expired - Fee Related US6013263A (en) 1993-06-07 1995-06-07 Measles virus peptides with antifusogenic and antiviral activities
US08/484,223 Expired - Fee Related US7794725B1 (en) 1993-06-07 1995-06-07 Isolated peptides derived from human immunodeficiency virus types 1 and 2 containing fusion inhibitory domains
US08/485,264 Expired - Fee Related US6228983B1 (en) 1993-06-07 1995-06-07 Human respiratory syncytial virus peptides with antifusogenic and antiviral activities
US08/919,597 Expired - Fee Related US6054265A (en) 1993-06-07 1997-09-26 Screening assays for compounds that inhibit membrane fusion-associated events
US10/267,748 Expired - Fee Related US7122190B2 (en) 1993-06-07 2002-10-08 Fusion proteins comprising DP-178 and other viral fusion inhibitor peptides useful for treating aids
US10/267,682 Expired - Fee Related US7273614B2 (en) 1993-06-07 2003-01-06 Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids
US10/739,355 Abandoned US20070037141A1 (en) 1994-06-07 2003-12-17 Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission
US11/253,274 Expired - Fee Related US7988974B2 (en) 1993-06-07 2005-10-17 Antifusogenic proteins comprising human immunodeficiency virus type 1 (HIV-1) gp41 DP-178 polypeptide variants and a macromolecular carrier

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/161,919 Abandoned US20110275146A1 (en) 1993-06-07 2011-06-16 Fusion proteins comprising dp-178 and other viral fusion inhibitor peptides useful for treating aids

Country Status (10)

Country Link
US (17) US6479055B1 (en)
EP (2) EP0793675B9 (en)
JP (3) JP2001523082A (en)
KR (1) KR100558087B1 (en)
AT (1) ATE308558T1 (en)
CA (1) CA2208420C (en)
DE (1) DE69534569T2 (en)
ES (1) ES2252747T3 (en)
NZ (1) NZ300002A (en)
WO (1) WO1996019495A1 (en)

Families Citing this family (121)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070202127A1 (en) * 1993-06-07 2007-08-30 Duke University Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids
US6479055B1 (en) 1993-06-07 2002-11-12 Trimeris, Inc. Methods for inhibition of membrane fusion-associated events, including respiratory syncytial virus transmission
EP0831873A4 (en) * 1995-06-07 2002-07-17 Trimeris Inc The treatment of hiv and other viral infections using combinatory therapy
US6271198B1 (en) 1996-11-06 2001-08-07 Genentech, Inc. Constrained helical peptides and methods of making same
FR2771011B1 (en) * 1997-11-17 2000-01-28 Hippocampe OBTAINING VACCINES TO PREVENT PATHOGENIC EFFECTS ASSOCIATED WITH RETROVIRAL INFECTION
US6281331B1 (en) 1998-03-23 2001-08-28 Trimeris, Inc. Methods and compositions for peptide synthesis
WO1999048513A1 (en) * 1998-03-23 1999-09-30 Trimeris, Inc. Methods and compositions for peptide synthesis
EP2261349A3 (en) 1998-05-01 2012-01-11 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
US20070026021A1 (en) * 1998-05-01 2007-02-01 Chiron S.R.I. Neisseria meningitidis antigens and compositions
US6656906B1 (en) 1998-05-20 2003-12-02 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
US6258782B1 (en) 1998-05-20 2001-07-10 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
US6797462B1 (en) 1998-06-23 2004-09-28 Uab Research Foundation Cell-based assay for immunodeficiency virus infectivity and sensitivity
FR2786773B1 (en) * 1998-12-03 2002-03-08 Univ Paris Curie ENVELOPE PROTEINS, METHODS AND USES
WO2000045833A1 (en) * 1999-02-02 2000-08-10 Uab Research Foundation, The Hiv drug resistance system
US7960540B2 (en) 1999-04-08 2011-06-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US20080318890A1 (en) * 1999-04-08 2008-12-25 Antisoma Research Limited Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
JP2002541264A (en) 1999-04-08 2002-12-03 ユーエイビー・リサーチ・ファウンデーション Anti-proliferative activity of G-rich oligonucleotide and its use for binding to nucleolin
US20080318889A1 (en) * 1999-04-08 2008-12-25 Antisoma Research Limited Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US8114850B2 (en) 1999-04-08 2012-02-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
JP4520441B2 (en) * 1999-05-13 2010-08-04 独立行政法人科学技術振興機構 Viral infection inhibitor targeting integrase N-terminal region
CN1351611A (en) * 1999-05-17 2002-05-29 康久化学公司 Long lasting fusion peptide inhibitors of viral infection
US6469136B1 (en) 1999-07-07 2002-10-22 Trimeris, Inc. Methods and composition for peptide synthesis
US6541020B1 (en) 1999-07-09 2003-04-01 Trimeris, Inc. Methods and compositions for administration of therapeutic reagents
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
US6706892B1 (en) 1999-09-07 2004-03-16 Conjuchem, Inc. Pulmonary delivery for bioconjugation
US7090851B1 (en) 1999-09-10 2006-08-15 Conjuchem Inc. Long lasting fusion peptide inhibitors of viral infection
WO2001024764A2 (en) * 1999-10-06 2001-04-12 The Trustees Of The University Of Pennsylvania Cell targeting compositions and methods of using the same
AU2001233340B2 (en) 2000-02-10 2006-05-04 Panacos Pharmaceuticals, Inc. Assay for detection of viral fusion inhibitors
US6623741B1 (en) * 2000-02-29 2003-09-23 Trimeris, Inc. Methods and compositions for inhibition of membrane fusion-associated events including RSV transmission
US6528308B1 (en) 2000-03-16 2003-03-04 Duke University Suppressor of HIV replication and transcription
EP2180064A3 (en) * 2000-10-18 2010-08-11 Pharmasset, Inc. Multiplex quantification of nucleic acids in diseased cells
WO2002044419A2 (en) * 2000-11-28 2002-06-06 Wyeth Expression analysis of kiaa nucleic acids and polypeptides useful in the diagnosis and treatment of prostate cancer
US6821731B2 (en) * 2000-11-28 2004-11-23 Wyeth Expression analysis of FKBP nucleic acids and polypeptides useful in the diagnosis of prostate cancer
EP1364053A2 (en) * 2000-11-28 2003-11-26 Wyeth Expression analysis of smarc nucleic acids and polypeptides useful in the diagnosis and treatment of prostate cancer
PT1479691E (en) * 2001-05-31 2007-02-28 John Erickson Long lasting fusion peptide inhibitors for hiv infection
CN1255548C (en) 2001-06-15 2006-05-10 霍夫曼-拉罗奇有限公司 Acetylation of GP41 framgnets
US7176278B2 (en) 2001-08-30 2007-02-13 Biorexis Technology, Inc. Modified transferrin fusion proteins
US20050227933A1 (en) * 2001-11-29 2005-10-13 Benkovic Stephen J Treatment of bacterial induced diseases using DNA methyl transferase inhibitors
EP2258712A3 (en) * 2002-03-15 2011-05-04 Multicell Immunotherapeutics, Inc. Compositions and Methods to Initiate or Enhance Antibody and Major-histocompatibility Class I or Class II-restricted T Cell Responses by Using Immunomodulatory, Non-coding RNA Motifs
AU2003277378A1 (en) * 2002-10-16 2004-05-04 Panacos Pharmaceuticals, Inc. Method for detecting viral inactivating agents
CA2443365C (en) * 2002-11-19 2010-01-12 F. Hoffmann-La Roche Ag Methods for the recombinant production of antifusogenic peptides
NZ567952A (en) * 2003-03-24 2009-12-24 Sequoia Pharmaceuticals Inc Long acting biologically active conjugates
CN1795274A (en) * 2003-03-26 2006-06-28 多单元免疫治疗公司 Selected rna motifs to include cell death and/or apoptosis
US20050281829A1 (en) * 2003-05-06 2005-12-22 Hehir Cristina A T Fc chimeric proteins with anti-HIV drugs
US7348004B2 (en) 2003-05-06 2008-03-25 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
HUE026384T2 (en) * 2003-05-06 2016-06-28 Biogen Hemophilia Inc Clotting factor chimeric proteins for treatment of a hemostatic disorder
US20050037947A1 (en) * 2003-05-06 2005-02-17 Bitonti Alan J. Inhibition of drug binding to serum albumin
WO2004108886A2 (en) * 2003-05-08 2004-12-16 Anderson Porter W Anti-hiv-1 compounds based upon a conserved amino acid sequence shared by gp160 and the human cd4 protein
CA2533878A1 (en) * 2003-07-29 2005-09-22 Immunomedics, Inc. Fluorinated carbohydrate conjugates
US20110104196A1 (en) * 2003-10-23 2011-05-05 Karp Nelson M Immunogenic composition and method of developing a vaccine based on fusion protein
US7611712B2 (en) * 2003-10-23 2009-11-03 Nelson M. Karp Immunogenic compositions capable of eliciting Th1 immune responses comprising an HIV-1 MA myristate binding site polypeptide
EP2261377B1 (en) * 2003-11-04 2013-10-02 The Administrators Of The Tulane Educational Fund Method of preventing virus:cell fusion by inhibiting the function of the fusion initiation region in RNA viruses having class I membrane fusogenic envelope proteins
US20080096809A1 (en) * 2003-12-22 2008-04-24 Yeda Research & Development Co. Ltd. Diastereomeric Peptides Useful as Inhibitors of Membrane Protein Assembly
JP5132939B2 (en) * 2003-12-23 2013-01-30 ネイションワイド チルドレンズ ホスピタル, インコーポレイテッド Haemophilusinfluenzae type IV pili
EP1696952A4 (en) 2003-12-23 2007-11-07 Centocor Inc Anti-retroviral agents, compositions, methods and uses
WO2005063800A2 (en) * 2003-12-31 2005-07-14 F. Hoffmann-La Roche Ag Peptide synthesis using decanting filter
EP1701972B1 (en) * 2003-12-31 2008-10-08 F. Hoffmann-La Roche Ag Process and systems for recovery of peptides
WO2005063792A2 (en) * 2003-12-31 2005-07-14 F. Hoffmann-La Roche Ag Methods for recovering cleaved peptide from a support
EP1701969B1 (en) * 2003-12-31 2007-10-24 F.Hoffmann-La Roche Ag Process for peptide synthesis using a reduced amount of deprotection agent
WO2005063799A2 (en) * 2003-12-31 2005-07-14 F.Hoffmann-La Roche Ag Peptide synthesis and deprotection with co-solvent
EP1708734A4 (en) * 2004-01-07 2009-06-17 Trimeris Inc HIV gp41 HR2-DERIVED SYNTHETIC PEPTIDES, AND THEIR USE IN THERAPY TO INHIBIT TRANSMISSION OF HUMAN IMMUNODEFICIENCY VIRUS
WO2005077103A2 (en) * 2004-02-12 2005-08-25 Regents Of The University Of Colorado Compositions and methods for modification and prevention of sars coronavirus infectivity
CA2565658A1 (en) * 2004-05-06 2005-11-17 Conjuchem Biotechnologies Inc. Compounds for specific viral target
GB0422439D0 (en) * 2004-10-08 2004-11-10 European Molecular Biology Lab Embl Inhibitors of infection
US20100093767A1 (en) * 2004-12-03 2010-04-15 Takeda San Diego, Inc. Mitotic Kinase Inhibitors
CA2592438C (en) * 2004-12-30 2013-09-03 F.Hoffmann-La Roche Ag Synthesis of peptide t-20 using peptide intermediate fragments
JP4886703B2 (en) * 2004-12-30 2012-02-29 エフ.ホフマン−ラ ロシュ アーゲー Synthesis of peptide T-1249 using peptide intermediate fragments
CN101115498A (en) * 2005-01-24 2008-01-30 耶达研究与发展有限公司 Hiv-1 gp41 fusion peptides for immunomodulation
WO2007022477A2 (en) * 2005-08-17 2007-02-22 Multicell Immunotherapeutics, Inc. Methods and compositions to generate and control the effector profile of t cells
US20070185025A1 (en) * 2005-09-11 2007-08-09 The Trustees Of Columbia University In The City Of New York Filoviral immunosuppressive peptides and uses thereof
US7456251B2 (en) 2006-02-02 2008-11-25 Trimeris, Inc. HIV fusion inhibitor peptides with improved biological properties
US8216592B2 (en) * 2006-04-19 2012-07-10 The Reed Institute Infection model for developing chemotherapeutic agents
US8222204B2 (en) * 2006-05-03 2012-07-17 The Administrators of the Tulane Educational Fund and Autoimmune Technologies, LLC Influenza inhibiting compositions and methods
CN101088557A (en) 2006-06-12 2007-12-19 天津市扶素生物技术有限公司 Medicine composition for preventing and treating HIV infection and its application
US8411779B2 (en) 2006-06-27 2013-04-02 Nec Corporation Communication system, transmitter, receiver and multiple access method
CA2658484A1 (en) * 2006-07-20 2008-01-24 Vical Incorporated Compositions and methods for vaccinating against hsv-2
TW200817438A (en) * 2006-08-17 2008-04-16 Hoffmann La Roche A conjugate of an antibody against CCR5 and an antifusogenic peptide
EP2054086A1 (en) 2006-08-17 2009-05-06 F. Hoffmann-Roche AG A conjugate of an antibody against ccr5 and an antifusogenic peptide
EP2479189B1 (en) 2006-12-12 2015-02-25 Biorexis Pharmaceutical Corporation Transferrin fusion protein libraries
US20090088378A1 (en) * 2007-01-12 2009-04-02 Omar Quraishi Long lasting inhibitors of viral infection
TW200902544A (en) * 2007-03-13 2009-01-16 Hoffmann La Roche Peptide-complement conjugates
KR20100016142A (en) * 2007-04-03 2010-02-12 트라이머리스, 인코퍼레이티드 Novel formulations for delivery of antiviral peptide therapeutics
EP2147016A2 (en) * 2007-05-16 2010-01-27 ConjuChem Biotechnologies Inc. Cysteic acid derivatives of anti-viral peptides
CL2008002092A1 (en) 2007-07-20 2009-05-29 Hoffmann La Roche Conjugate containing two or more antifusogenic peptides and an anti-cd-4 antibody; Method of production; pharmaceutical composition comprising it; antifusogenic polypeptides and use of the conjugate to treat viral infections.
MX2010003179A (en) * 2007-09-25 2010-04-30 Trimeris Inc Methods of synthesis for therapeuthic anti-hiv peptides.
US20090131351A1 (en) * 2007-11-16 2009-05-21 Antisoma Research Limited Methods, compositions, and kits for modulating tumor cell proliferation
US7892562B2 (en) * 2008-10-08 2011-02-22 Karp Nelson M Methods of inducing TH-1 immune responses to HIV-1 by administering UV/psoralen-treated desialated inactiviated HIV-1 virions deficient in CD55 and CD59
EP2442829B1 (en) 2009-06-18 2021-02-24 Dana-Farber Cancer Institute, Inc. Structured viral peptide compositions and methods of use
EP2528623A2 (en) 2010-01-26 2012-12-05 The Regents of the University of Colorado, a body corporate Respiratory virus epitopes templated into double stranded coiled -coils and use thereof in immunization
WO2011133948A2 (en) 2010-04-22 2011-10-27 Longevity Biotech, Inc. Highly active polypeptides and methods of making and using the same
KR20130045385A (en) 2010-09-14 2013-05-03 에프. 호프만-라 로슈 아게 Serpin-finger fusion polypeptide
EP2447277A1 (en) 2010-10-28 2012-05-02 Laboratorios Del. Dr. Esteve, S.A. Vaccine compositions based on modified gp41 immunogens
WO2012135831A1 (en) 2011-04-01 2012-10-04 Yale University Cell-penetrating anti-dna antibodies and uses thereof inhibit dna repair
US20130172536A1 (en) * 2011-08-16 2013-07-04 Shenzhen Weiguang Biological Products Co.,Ltd. Intravenous Cytomegalovirus Human Immune Globulin and Manufacturing Method Thereof
US9951349B2 (en) 2011-09-27 2018-04-24 Yale University Compositions and methods for transient expression of recombinant RNA
CA2794397A1 (en) * 2011-10-27 2013-04-27 Institut National De La Recherche Scientifique Modulation of ul24 interactions with protein targets and uses thereof for inhibition of herpesvirus infection
EP2834264A1 (en) 2012-04-04 2015-02-11 Yeda Research and Development Co. Ltd. Lipopeptide conjugates comprising sphingolipid and hiv gp41 derived peptides
WO2014016152A1 (en) 2012-07-27 2014-01-30 Institut National De La Sante Et De La Recherche Medicale Cd147 as receptor for pilus-mediated adhesion of meningococci to vascular endothelia
KR101861416B1 (en) * 2012-09-13 2018-05-25 유니베르시떼 드 제네브 Cell penetrating peptides
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US9649375B2 (en) * 2013-03-14 2017-05-16 The Administrators Of The Tulane Educational Fund Immunogenic peptide conjugate and method for inducing an anti-influenza therapeutic antibody response therewith
SG10201913874TA (en) 2013-03-15 2020-03-30 Biogen Ma Inc Factor ix polypeptide formulations
US9789164B2 (en) 2013-03-15 2017-10-17 Longevity Biotech, Inc. Peptides comprising non-natural amino acids and methods of making and using the same
US9341391B2 (en) * 2013-12-31 2016-05-17 Bollente Companies, Inc. Automatically controlled flow-through water heater system
US10040867B2 (en) 2014-03-04 2018-08-07 Yale University Cell penetrating anti-guanosine antibody based therapy for cancers with Ras mutations
US10238742B2 (en) 2014-06-25 2019-03-26 Yale University Cell penetrating nucleolytic antibody based cancer therapy
CA2954920A1 (en) * 2014-07-14 2016-01-21 The Regents Of The University Of California A protein tagging system for in vivo single molecule imaging and control of gene transcription
CN105989283B (en) 2015-02-06 2019-08-09 阿里巴巴集团控股有限公司 A kind of method and device identifying virus mutation
JP7189021B2 (en) * 2016-03-16 2022-12-13 アマル セラピューティクス エスエー Combination of an immune checkpoint modulator and a conjugate comprising a cell penetrating peptide, a cargo and a TLR peptide agonist for use in medicine
JP2019518040A (en) 2016-06-15 2019-06-27 イェール ユニバーシティーYale University Antibody-Mediated Autocatalytic Delivery of Nanocarriers Targeted to Tumors
IL265479B (en) 2016-09-21 2022-09-01 Amal Therapeutics Sa Fusion comprising a cell penetrating peptide, a multi epitope and a tlr peptide agonist for treatment of cancer
WO2018059214A1 (en) * 2016-09-29 2018-04-05 广州君赫生物科技有限公司 Compounds affecting saicar synthesis, and applications
WO2018192323A1 (en) 2017-04-20 2018-10-25 广州君赫生物科技有限公司 Applications of spermidine and its derivative
EP3539562A1 (en) 2018-03-12 2019-09-18 Eberhard Karls Universität Tübingen Medizinische Fakultät Immunotherapeutic peptides
CN109207504B (en) * 2018-09-26 2022-03-18 四川大学 Preparation method of lactobacillus salivarius for effectively immunizing IB and ND
JP7333538B2 (en) * 2018-10-09 2023-08-25 東亞合成株式会社 Antiviral peptide and its use
CN109320594B (en) * 2018-11-13 2021-09-28 四川大学 Virus-like particle for avian infectious bronchitis and newcastle disease, preparation method and application
CN112029735B (en) * 2020-08-31 2022-04-12 中国农业科学院兰州兽医研究所 Foot-and-mouth disease virus non-structural protein 3B dominant epitope deletion marker strain and preparation method and application thereof
CN112410351A (en) * 2020-11-12 2021-02-26 山东农业大学 Duplex attenuated vaccine for resisting cucumber mosaic virus and potato virus X and application thereof
CN112410312A (en) * 2020-11-27 2021-02-26 江南大学 Cyclohexanone monooxygenase and application thereof

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4609546A (en) * 1982-06-24 1986-09-02 Japan Chemical Research Co., Ltd. Long-acting composition
US4659669A (en) * 1981-03-02 1987-04-21 Regents Of The Univ. Of California Microbial expression of human influenza hemagglutinin proteins
US4707358A (en) * 1984-01-30 1987-11-17 The University Of Chicago Vaccine against Epstein-Barr Virus
US4761470A (en) * 1985-12-16 1988-08-02 Merck & Co., Inc. Immunogenic synthetic peptide capable of eliciting herpes simplex virus neutralizing antibody
US5057211A (en) * 1988-12-19 1991-10-15 Baummer George P Benefication apparatus and process for land and seabed mining
US5116725A (en) * 1984-08-08 1992-05-26 Scripps Clinic And Research Foundation Assay for Epstein-Barr virus infection with solid phase bound synthetic polypeptides
US5141867A (en) * 1987-02-02 1992-08-25 E. I. Du Pont De Nemours And Company Nucleotide sequence encoding a human immunodeficiency virus antigen
US5156949A (en) * 1984-10-31 1992-10-20 Chiron Corporation Immunoassays for antibody to human immunodeficiency virus using recombinant antigens
US5268358A (en) * 1988-12-08 1993-12-07 Cor Therapeutics, Inc. PDGF receptor blocking peptides
US5444044A (en) * 1992-03-26 1995-08-22 New York Blood Center Synthetic polypeptides as inhibitors of HIV-1
US5464933A (en) * 1993-06-07 1995-11-07 Duke University Synthetic peptide inhibitors of HIV transmission
US5656480A (en) * 1992-07-20 1997-08-12 Duke University Compounds which inhibit HIV replication
US5853978A (en) * 1985-12-04 1998-12-29 Genentech, Inc. Molecularly cloned acquired immunodeficiency syndrome polypeptides and methods of use
US5876969A (en) * 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US6001977A (en) * 1984-08-22 1999-12-14 The United States Of America As Represented By The Department Of Health And Human Services Cloning and expression of HTLV-III DNA
US6017536A (en) * 1993-06-07 2000-01-25 Trimeris, Inc. Simian immunodeficiency virus peptides with antifusogenic and antiviral activities
US6025325A (en) * 1995-05-05 2000-02-15 Hoffman-La Roche Inc. Pegylated obese (ob) protein compositions
US6054265A (en) * 1993-06-07 2000-04-25 Trimeris, Inc. Screening assays for compounds that inhibit membrane fusion-associated events
US6248574B1 (en) * 1989-12-13 2001-06-19 Avigdor Shaffermann Polypeptides selectively reactive with antibodies against human immunodeficiency virus and vaccines comprising the polypeptides
US6261564B1 (en) * 1984-10-18 2001-07-17 Institut Pasteur Peptides of human immunodeficiency virus type 1 (HIV-1)
US6440657B1 (en) * 1983-12-05 2002-08-27 Institut Pasteur Nucleic acids and peptides of human immunodeficiency virus type (HIV-1)
US20070202127A1 (en) * 1993-06-07 2007-08-30 Duke University Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4859765A (en) * 1983-10-17 1989-08-22 Syntex (U.S.A.) Inc. Synthetic peptide sequences useful in biological and pharmaceutical applications and methods of manufacture
IL76082A (en) 1984-08-22 1991-07-18 Us Health Molecular clones of the genome of htlv-iii and a process for the preparation thereof
GB8501473D0 (en) 1985-01-21 1985-02-20 Pasteur Institut Cloned dna sequences
ES8705522A1 (en) 1984-10-18 1987-05-01 Pasteur Institut Gag antigen and its use for the detection of lav infection and in immunogenic compositions
EP0187041B1 (en) 1984-12-24 1996-05-15 Genentech, Inc. Fusions of AIDS-related polypeptides
WO1986006414A1 (en) 1985-04-29 1986-11-06 Genetic Systems Corporation Synthetic antigens for the detection of aids-related disease
US5075211A (en) * 1986-03-26 1991-12-24 Genetic Systems Corporation Synthetic antigen for the detection of AIDS-related disease
WO1988005051A1 (en) 1986-12-30 1988-07-14 United States Of America, Represented By The Unite Synthetic peptides which induce cellular immunity of the aids virus and aids viral proteins
ES2104556T3 (en) 1987-01-16 1997-10-16 Pasteur Institut PEPTIDES THAT HAVE IMMUNOLOGICAL PROPERTIES OF HIV-2.
NO881151L (en) 1987-03-27 1988-09-28 Syntello Ab SYNTHETIC HIV-1 ANTIGEN.
AU1711888A (en) 1987-04-24 1988-12-02 Biogen, Inc. Immunotherapeutic methods and compositions
US5223254A (en) * 1987-09-29 1993-06-29 Praxis Biologics, Inc. Respiratory syncytial virus: vaccines
EP0323157A3 (en) 1987-12-24 1990-07-25 The University Of Melbourne Antiviral compounds and methods
US5241047A (en) 1988-12-08 1993-08-31 Biochem Pharma Inc. Synthetic peptides and mixtures thereof for detecting HIV antibodies
AU608413B2 (en) 1988-03-30 1991-03-28 Abbott Laboratories Mouse monoclonal antibody (5-21-3) to human immunodeficiency virus gp41 protein
DE68915165T2 (en) * 1988-04-22 1994-09-08 Upjohn Co IMMUNOGENIC SECTIONS OF HUMAN PARAIN INFLUENCE VIRUS TYPE 3 CONTAINING CHIMERIC GLYCOPROTEINS.
EP0362909A3 (en) 1988-08-26 1990-11-14 Akzo N.V. Synthetic polypeptides immunochemically reactive with hiv-antibodies
EP0362910A3 (en) 1988-08-27 1991-01-09 Akzo N.V. Synthetic peptides immunochemically reactive with hiv-antibodies
EP0362927A3 (en) 1988-10-06 1990-11-14 Akzo N.V. Synthetic polypeptides immunochemically reactive with hiv antibodies
DE68925909T2 (en) 1988-12-20 1996-11-14 Clarity Technologies Inc SYNTHETIC HIV-LIKE PEPTIDES, THEIR COMPOSITIONS AND USES
FR2677346B1 (en) * 1991-06-10 1993-08-27 Atochem PROCESS FOR PURIFYING AN AQUEOUS SOLUTION OF ALKALI METAL CHLORIDE BY REMOVING AMMONIUM AND IODINE.
IL102092A (en) 1991-06-11 1996-11-14 Microgenesys Inc Use of recombinant hiv envelope protein in medicament for treating hiv and therapeutic composition containing the agglomerated protein
GB9120221D0 (en) * 1991-09-23 1991-11-06 Smithkline Beecham Biolog Novel compounds
GB9200117D0 (en) 1992-01-06 1992-02-26 Connaught Lab Production of recombinant chimeric proteins for vaccine use
EP0831873A4 (en) * 1995-06-07 2002-07-17 Trimeris Inc The treatment of hiv and other viral infections using combinatory therapy
US6156949A (en) * 1998-12-28 2000-12-05 Mobil Oil Corporation Selective pseudocumene production by xylene methylation

Patent Citations (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4659669A (en) * 1981-03-02 1987-04-21 Regents Of The Univ. Of California Microbial expression of human influenza hemagglutinin proteins
US4609546A (en) * 1982-06-24 1986-09-02 Japan Chemical Research Co., Ltd. Long-acting composition
US6440657B1 (en) * 1983-12-05 2002-08-27 Institut Pasteur Nucleic acids and peptides of human immunodeficiency virus type (HIV-1)
US4707358A (en) * 1984-01-30 1987-11-17 The University Of Chicago Vaccine against Epstein-Barr Virus
US5116725A (en) * 1984-08-08 1992-05-26 Scripps Clinic And Research Foundation Assay for Epstein-Barr virus infection with solid phase bound synthetic polypeptides
US6001977A (en) * 1984-08-22 1999-12-14 The United States Of America As Represented By The Department Of Health And Human Services Cloning and expression of HTLV-III DNA
US6610476B1 (en) * 1984-08-22 2003-08-26 The United States Of America As Represented By The Department Of Health And Human Services Detection of HIV-1 DNA
US6858712B1 (en) * 1984-08-22 2005-02-22 The United States Of America As Represented By The Department Of Health And Human Services Cloning and expression of HTLV-III DNA
US6261564B1 (en) * 1984-10-18 2001-07-17 Institut Pasteur Peptides of human immunodeficiency virus type 1 (HIV-1)
US5156949A (en) * 1984-10-31 1992-10-20 Chiron Corporation Immunoassays for antibody to human immunodeficiency virus using recombinant antigens
US6531276B1 (en) * 1984-10-31 2003-03-11 Chiron Corporation Methods for detecting human immunodeficiency virus nucleic acid
US5853978A (en) * 1985-12-04 1998-12-29 Genentech, Inc. Molecularly cloned acquired immunodeficiency syndrome polypeptides and methods of use
US4761470A (en) * 1985-12-16 1988-08-02 Merck & Co., Inc. Immunogenic synthetic peptide capable of eliciting herpes simplex virus neutralizing antibody
US5141867A (en) * 1987-02-02 1992-08-25 E. I. Du Pont De Nemours And Company Nucleotide sequence encoding a human immunodeficiency virus antigen
US5268358A (en) * 1988-12-08 1993-12-07 Cor Therapeutics, Inc. PDGF receptor blocking peptides
US5057211A (en) * 1988-12-19 1991-10-15 Baummer George P Benefication apparatus and process for land and seabed mining
US6248574B1 (en) * 1989-12-13 2001-06-19 Avigdor Shaffermann Polypeptides selectively reactive with antibodies against human immunodeficiency virus and vaccines comprising the polypeptides
US5876969A (en) * 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US5840843A (en) * 1992-03-26 1998-11-24 The New York Blood Center Synthetic polypeptides as inhibitors of HIV-1
US5444044A (en) * 1992-03-26 1995-08-22 New York Blood Center Synthetic polypeptides as inhibitors of HIV-1
US6573078B1 (en) * 1992-07-20 2003-06-03 Duke University Compounds which inhibit HIV replication
US5656480A (en) * 1992-07-20 1997-08-12 Duke University Compounds which inhibit HIV replication
US6479055B1 (en) * 1993-06-07 2002-11-12 Trimeris, Inc. Methods for inhibition of membrane fusion-associated events, including respiratory syncytial virus transmission
US6440656B1 (en) * 1993-06-07 2002-08-27 Trimeris, Inc. Methods for the inhibition of respiratory syncytial virus transmission
US6133418A (en) * 1993-06-07 2000-10-17 Duke University Synthetic peptide inhibitors of HIV transmission
US6017536A (en) * 1993-06-07 2000-01-25 Trimeris, Inc. Simian immunodeficiency virus peptides with antifusogenic and antiviral activities
US6054265A (en) * 1993-06-07 2000-04-25 Trimeris, Inc. Screening assays for compounds that inhibit membrane fusion-associated events
US5464933A (en) * 1993-06-07 1995-11-07 Duke University Synthetic peptide inhibitors of HIV transmission
US7122190B2 (en) * 1993-06-07 2006-10-17 Duke University Fusion proteins comprising DP-178 and other viral fusion inhibitor peptides useful for treating aids
US20070202127A1 (en) * 1993-06-07 2007-08-30 Duke University Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids
US20070202123A1 (en) * 1993-06-07 2007-08-30 Duke University Fusion proteins comprising DP-178 and other viral fusion inhibitor peptides useful for treating aids
US7273614B2 (en) * 1993-06-07 2007-09-25 Duke University Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids
US7794725B1 (en) * 1993-06-07 2010-09-14 Trimeris, Inc. Isolated peptides derived from human immunodeficiency virus types 1 and 2 containing fusion inhibitory domains
US20070037141A1 (en) * 1994-06-07 2007-02-15 Duke University Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission
US6025325A (en) * 1995-05-05 2000-02-15 Hoffman-La Roche Inc. Pegylated obese (ob) protein compositions

Also Published As

Publication number Publication date
US6228983B1 (en) 2001-05-08
AU4473496A (en) 1996-07-10
US20070202123A1 (en) 2007-08-30
US6054265A (en) 2000-04-25
US6093794A (en) 2000-07-25
US20070037141A1 (en) 2007-02-15
CA2208420A1 (en) 1996-06-27
US20040052820A1 (en) 2004-03-18
EP0793675B9 (en) 2007-05-09
US6824783B1 (en) 2004-11-30
JP2009213475A (en) 2009-09-24
US6013263A (en) 2000-01-11
US6479055B1 (en) 2002-11-12
US6068973A (en) 2000-05-30
US7988974B2 (en) 2011-08-02
EP1714974A3 (en) 2007-03-21
US7273614B2 (en) 2007-09-25
EP1714974A2 (en) 2006-10-25
US7794725B1 (en) 2010-09-14
US6333395B1 (en) 2001-12-25
US20040033235A1 (en) 2004-02-19
CA2208420C (en) 2010-11-09
NZ300002A (en) 2000-02-28
EP0793675A4 (en) 1998-08-19
JP2001523082A (en) 2001-11-20
US6951717B1 (en) 2005-10-04
US6060065A (en) 2000-05-09
DE69534569D1 (en) 2005-12-08
EP0793675A1 (en) 1997-09-10
KR100558087B1 (en) 2007-01-31
ES2252747T3 (en) 2006-05-16
JP2006176529A (en) 2006-07-06
EP0793675B1 (en) 2005-11-02
US20110275146A1 (en) 2011-11-10
ATE308558T1 (en) 2005-11-15
KR987000333A (en) 1998-03-30
US7122190B2 (en) 2006-10-17
AU714695B2 (en) 2000-01-06
WO1996019495A1 (en) 1996-06-27
DE69534569T2 (en) 2006-08-10

Similar Documents

Publication Publication Date Title
US7794725B1 (en) Isolated peptides derived from human immunodeficiency virus types 1 and 2 containing fusion inhibitory domains
WO1996019495A9 (en) Methods and compositions for inhibition of membrane fusion-associated events, including hiv transmission
EP0774971B1 (en) Synthetic peptide inhibitors of hiv transmission
US6750008B1 (en) Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission
US6623741B1 (en) Methods and compositions for inhibition of membrane fusion-associated events including RSV transmission
US20070202127A1 (en) Nucleic acids encoding DP-178 and other viral fusion inhibitor peptides useful for treating aids
AU714695C (en) Methods and compositions for inhibition of membrane fusion-associated events, including HIV transmission

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION