PEPTIDE VIRAL ENTRY INHIBITORS

-

The present invention provides, inter alia, peptide compositions and methods for treating and preventing Flaviviridae virus (e.g., HCV) infections.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

This application is a divisional of U.S. patent application Ser. No. 11/264,509, filed Nov. 1, 2005 which claims the benefit of U.S. provisional patent application No. 60/624,204, filed Nov. 2, 2004; each of which is herein incorporated by reference in its entirety.

FIELD OF THE INVENTION

The present invention comprises compositions and methods for treating or preventing a viral infection in subject.

BACKGROUND OF THE INVENTION

Approximately 170 million people are infected with hepatitis C virus (HCV) world-wide. Current therapies are helpful, but are not effective in every patient. Moreover, many of the current therapies result in unwanted side-effects.

HCV particles attach and enter target cells through interactions between their viral glycoproteins E1 and E2 and cell surface receptor molecules. The viral entry step has been difficult to study because there is no reliable, easy to detect system for performing HCV/tissue culture infection. Hsu et al. (Proc. Natl. Acad. Sci. USA. 100(12):7271-7276 (2003)) and Bartosch et al. (J. Exp. Med. 197(5):633-42 (2003)) reported the generation of HCV pseudoparticles that use the core proteins of HIV-1 and authentic HCV E1 and E2 proteins. The pseudoparticles specifically infect hepatocytes and liver-derived cell lines. Such pseudoparticle infection can be neutralized by HCV patient serum as well as antibodies against E1 and E2 glycoproteins. CD81 is a host cell protein required, but not sufficient, for HCV pseudoparticle entry.

Peptides derived from HIV-1 envelope glycoproteins can bind HIV-1 and inhibit infection at the cell fusion step (Baldwin et al., Curr. Med. Chem. 10(17): 1633-1642 (2003)). Foung et al. (U.S. Pat. No. 6,692,908) discuss use of antibodies which inhibit HCV E1 and E1 binding to CD81 for treatment of HCV infection. Garry et al. (WO 2004/044220) discuss use of the peptides from the E1 envelope glycoprotein of hepaciviruses and E2 envelope glycoprotein of pestiviruses for treatment of viral infection. Furthermore, VanCompernolle et al. disclose imidazole-based small molecules that inhibit E2/CD81 binding (Virology 314(1):371-380 (2003)).

As mentioned above, in spite of the current availability of treatments for HCV, infections in many patients are not responsive. There remains a need in the art for additional treatments to the prevention and treatment of HCV infection.

SUMMARY OF THE INVENTION

The present invention, inter alia, meets the need in the art for additional therapies for the treatment and prevention of HCV infection.

The present invention provides an isolated polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139. In an embodiment of the invention, the polypeptide comprises or consists of the amino acid sequence of SEQ ID NO: 3, 7, 9, 10, 12-28, 30-38, 40, 42, 44-60, 62-68, 70, 71, 73-103, 106-120, 121-130, 132-138.

Another embodiment of the invention comprises an isolated polypeptide (i) comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 82-84, 87-97, 99, 101-103 and 117-138; or (ii) consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-81, 85, 86, 98, 100, 104-116 and 139. A further embodiment of the invention comprises an isolated polypeptide (i) comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 82-84, 87-97, 99, 101-103, 117-130 and 132-138; or (ii) consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3, 7, 9, 10, 12-28, 30-38, 40, 42, 44-60, 62-68, 70-81, 85, 86, 98, 100 and 106-116.

Another embodiment of the invention comprises a pharmaceutical composition comprising an isolated polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139 and a pharmaceutically acceptable carrier. A further embodiment of the invention comprises a composition comprising an isolated polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139 in association with one or more members selected from the group consisting of anti-human CD81 antibody, ribavirin, interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, and pegylated consensus interferon (e.g., a kit). In an embodiment of the invention, an isolated peptide of the invention is combined in association with an HCV protease inhibitor or an HCV polymerase inhibitor.

The present invention also includes an oligonucleotide comprising the nucleotide sequence set forth in SEQ ID NO: 140 or 141.

The present invention also provides an isolated polynucleotide encoding a polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139. In an embodiment of the invention, the polynucleotide encodes a polypeptide comprising or consisting of the amino acid sequence of SEQ ID NO: 3, 7, 9, 10, 12-28, 30-38, 40, 42, 44-60, 62-68, 70, 71, 73-103, 106-120, 121-130 or 132-138. A further embodiment of the invention comprises a recombinant vector comprising an isolated polynucleotide encoding a polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139. Another embodiment of the invention comprises an isolated host cell comprising the vector.

The present invention also provides a method for making a polypeptide (e.g., any of SEQ ID NOs: 3-139) comprising culturing a host cell comprising a polynucleotide encoding the polypeptide under conditions in which the polynucleotide is expressed and, optionally, isolating the polypeptide from the culture.

The present invention also provides a method for inhibiting entry of a virus which is a member of the Flaviviridae family (e.g., hepatitis C virus) into a cell (e.g., in vitro or in vivo) comprising contacting the cell with a polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139. Also provided by the present invention is a method for treating or preventing infection of a subject with a virus which is a member of the Flaviviridae family comprising administering to said subject a therapeutically effective amount of a polypeptide of the present invention (e.g., any of SEQ ID NOs3-139). In an embodiment of the invention, the polypeptide (e.g., any of SEQ ID NOs: 3-139) is administered in association with one or more members selected from the group consisting of anti-human CD81 antibody, ribavirin, interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, and pegylated consensus interferon. In an embodiment of the invention, the pegylated interferon alfa that is administered is a pegylated interferon alfa-2b and wherein the amount of pegylated interferon alfa-2b that is administered in the treatment time period is about 0.5 to 1.5 micrograms per kilogram body weight of pegylated interferon alfa-2b protein per week on a weekly basis for at least twenty-four weeks (e.g., about 48 weeks). In an embodiment of the invention, said polypeptide (e.g., any of SEQ ID NOs: 3-139) is administered for a treatment time period sufficient to eradicate detectable hepatitis C virus-RNA and to maintain no detectable hepatitis C virus RNA for at least twelve weeks after the end of the treatment time period. In a further embodiment of the invention, said polypeptide (e.g., any of SEQ ID NOs: 3-139) is administered in association with a therapeutically effective amount of an interferon for a treatment time period sufficient to eradicate detectable hepatitis C virus-RNA and to maintain no detectable hepatitis C virus RNA for at least twelve weeks after the end of the treatment time period. In an embodiment of the invention, the host is infected with multiple hepatitis C virus genotypes (e.g., hepatitis C virus genotype 1 and/or hepatitis C virus genotype 2 and/or hepatitis C virus genotype 3).

The present invention also provides a method for preventing infection of a host, with a virus which is a member of the Flaviviridae family of viruses (e.g., hepatitis C virus), following transplantation of a liver into said host or transfusion of blood into said host comprising administering (e.g., parenterally (e.g., intramuscularly, intravenously, subcutaneously) or non-parenterally (e.g., orally)) to said host (e.g., before, during or after said transplantation or transfusion) a therapeutically effective amount of a polypeptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-139. In an embodiment of the invention, the polypeptide (e.g., any of SEQ ID NOs: 3-139) is administered in association with anti-human CD81 antibody, interferon-alfa, pegylated interferon-alfa or albumin-interferon-alpha (e.g., interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, or pegylated consensus interferon). In an embodiment of the invention, the polypeptide (e.g., any of SEQ ID NOs: 3-139) is administered in association with ribavirin.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides peptide compositions (e.g., any of SEQ ID NOs: 3-139) and methods for treating or preventing an infection by a virus which is a member of the Flaviviridae family (e.g., HCV) in a subject (e.g., a patient who has received a liver transplant) by administering one or more of the peptides to the subject. In an embodiment of the invention, the peptide comprises the amino acid sequence of any of SEQ ID NOs: 3, 7, 9, 10, 12-28, 30-38, 40, 42, 44-60, 62-68, 70, 71, 73-103, 106-120, 121-130, 132-138.

A patient or host suffering from an infection by a Flaviviridae virus, such as HCV (e.g., a chronic or acute HCV infection), can be treated by administering to the patient an E1-E2 polypeptide (e.g., any of SEQ ID NOs: 3-139) or a pharmaceutically acceptable salt thereof.

An E1-E2 peptide (e.g., comprising the amino acid sequence of any of SEQ ID NO: 3-139) can also be administered to a patient in association with one or more other anti-viral agents such as an anti-human CD81 antibody, pegylated or unpegylated interferon alfa-2a, pegylated or unpegylated interferon alfa-2b, pegylated or unpegylated interferon alfa-2c, pegylated or unpegylated interferon alfa n-1, pegylated or unpegylated interferon alfa n-3 or pegylated or unpegylated consensus interferon.

In a liver transplantation procedure, the donor liver can come from a living donor (i.e., living donor liver transplantation (LDLT)) wherein a portion of the donor's liver is removed and introduced into the recipient. Alternatively, the transplant can be from a deceased donor wherein the entire liver is removed and transplanted.

For example, the present invention includes, but is not limited to methods for treating or preventing infections caused by members of the Hepacivirus genus which includes the hepatitis C virus (HCV). HCV includes several types, subtypes and isolates:

hepatitis C virus (isolate 1)

hepatitis C virus (isolate BK)

hepatitis C virus (isolate EC1)

hepatitis C virus (isolate EC10)

hepatitis C virus (isolate HC-J2)

hepatitis C virus (isolate HC-J5)

hepatitis C virus (isolate HC-J6)

hepatitis C virus (isolate HC-J7)

hepatitis C virus (isolate HC-J8)

hepatitis C virus (isolate HC-JT)

hepatitis C virus (isolate HCT18)

hepatitis C virus (isolate HCT27)

hepatitis C virus (isolate HCV-476)

hepatitis C virus (isolate HCV-KF)

hepatitis C virus (isolate Hunan)

hepatitis C virus (isolate Japanese)

hepatitis C virus (isolate Taiwan)

hepatitis C virus (isolate TH)

hepatitis C virus isolate H

hepatitis C virus type 1

    • hepatitis C virus type 1a
      • hepatitis C virus strain H77
    • hepatitis C virus type 1b
    • hepatitis C virus type 1c
    • hepatitis C virus type 1d
    • hepatitis C virus type 1e
    • hepatitis C virus type 1f

hepatitis C virus type 10

hepatitis C virus type 2

    • hepatitis C virus type 2a
    • hepatitis C virus type 2b
    • hepatitis C virus type 2c
    • hepatitis C virus type 2d
    • hepatitis C virus type 2f

hepatitis C virus type 3

    • hepatitis C virus type 3a
    • hepatitis C virus type 3b
    • hepatitis C virus type 3g

hepatitis C virus type 4

    • hepatitis C virus type 4a
    • hepatitis C virus type 4c
    • hepatitis C virus type 4d
    • hepatitis C virus type 4f
    • hepatitis C virus type 4h
    • hepatitis C virus type 4k

hepatitis C virus type 5

    • hepatitis C virus type 5a

hepatitis C virus type 6

    • hepatitis C virus type 6a

hepatitis C virus type 7

    • hepatitis C virus type 7a
    • hepatitis C virus type 7b

hepatitis C virus type 8

    • hepatitis C virus type 8a

The present invention also includes methods for treating or preventing infection caused by members of the Flavivirus genus. The Flavivirus genus includes Yellow fever virus; Tick-borne viruses such as the Gadgets Gully virus, Kadam virus, Kyasanur Forest disease virus, Langat virus, Omsk hemorrhagic fever virus, Powassan virus, Royal Farm virus, Karshi virus, Tick-borne encephalitis virus, Neudoerfl virus, Neudoerfl virus, Sofjin virus, Louping ill virus and the Negishi virus; seabird tick-borne viruses such as the Meaban virus, Saumarez Reef virus, and the Tyuleniy virus; mosquito-borne viruses such as the Aroa virus, Bussuquara virus, Iguape virus and the Naranjal virus; Dengue viruses such as the Dengue virus and the Kedougou virus; Japanese encephalitis viruses such as the Cacipacore virus, Koutango virus, Japanese encephalitis virus, Murray Valley encephalitis virus, Alfuy virus, St. Louis encephalitis virus, Usutu virus, West Nile virus, Kunjin virus and the Yaounde virus; Kokobera viruses such as the Kokobera virus and the Stratford virus; Ntaya viruses such as the Bagaza virus, Ilheus virus, Rocio virus, Israel turkey meningoencephalomyelitis virus, Ntaya virus and the Tembusu virus; Spondweni viruses such as the Zika virus and the Spondweni virus; Yellow fever viruses such as the Banzi virus, Bouboui virus, Edge Hill virus, Jugra virus, Saboya virus, Potiskum virus, Sepik virus, Uganda S virus, Wesselsbron virus and the Yellow fever virus; Entebbe viruses such as the Entebbe bat virus, Sokoluk virus, and the Yokose virus; Modoc viruses such as the Apoi virus, Cowbone Ridge virus, Jutiapa virus, Modoc virus, Sal Vieja virus and the San Perlita virus; Rio Bravo viruses such as the Bukalasa bat virus, Carey Island virus, Dakar bat virus, Montana myotis leukoencephalitis virus, Phnom Penh bat virus, Batu Cave virus, Rio Bravo virus, Tamana bat virus, and the Cell fusing agent virus.

The present invention also includes methods for treating or preventing infection caused by members of the Pestivirus genus. The Pestivirus genus includes Bovine viral diarrhea virus 1, Border disease virus (sheep), Bovine viral diarrhea virus 1, Bovine viral diarrhea virus 2, Classical swine fever virus, and Hog cholera virus.

Moreover, the present invention includes methods for treating or preventing infections caused by Hepatitis G virus or Hepatitis GB virus-A, B or C.

A “host”, “subject” or “patient” may be any organism, such as a mammal (e.g., primate, dog, cat, cow, horse, pig, goat, rat, mouse, bird), preferably a human. A host, subject or patient can be an organism, such as a human, that is co-infected with another virus such as the human immunodeficiency virus (HIV; e.g., HIV-1 or HIV-2). Accordingly, the present invention comprises methods and compositions for treating Flaviviridae infection in a host this also infected with HIV.

Molecular Biology

In accordance with the present invention there may be employed conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (herein “Sambrook, et al., 1989”); DNA Cloning: A Practical Approach, Volumes I and II (D. N. Glover ed. 1985); Oligonucleotide Synthesis (M. J. Gait ed. 1984); Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. (1985)); Transcription And Translation (B. D. Hames & S. J. Higgins, eds. (1984)); Animal Cell Culture (R. I. Freshney, ed. (1986)); Immobilized Cells And Enzymes (IRL Press, (1986)); B. Perbal, A Practical Guide To Molecular Cloning (1984); F. M. Ausubel, et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (1994).

The HCV E1 protein is well known in the art. For example, the an amino acid sequence of the HCV E1 protein is:

(SEQ ID NO: 1) yqvrnssgly hvtndcpnss ivyeaadail htpgcvpcvr egnasrcwva vtptvatrdg klpttqlrrh idllvgsatl csalyvgdlc gsvflvgqlf tfsprrhwtt qdcncsiypg hitghrmawd mmmnwsptaa lvvaqllrip qaimdmiaga hwgvlagiay fsmvgnwakv lvvlllfagv da

(see also Genbank accession no. NP 751920)

The HCV E2 protein is well known in the art. For example, the an amino acid sequence of the HCV E2 protein is:

(SEQ ID NO: 2) ethvtggsag rttaglvgll tpgakqniql intngswhin stalncnesl ntgwlaglfy qhkfnssgcp erlascrrlt dfaqgwgpis yangsglder pycwhypprp cgivpaksvc gpvycftpsp vvvgttdrsg aptyswgand tdvfvlnntr pplgnwfgct wmnstgftkv cgappcvigg vgnntllcpt dcfrkhpeat ysrcgsgpwi tprcmvdypy rlwhypctin ytifkvrmyv ggvehrleaa cnwtrgercd ledrdrsels plllsttqwq vlpcsfttlp alstglihlh qnivdvqyly gvgssiaswa ikweyvvllf llladarvcs clwmmllisq aea

(see also Genbank accession nos. NP751921; AAB67038; AAB67036 and P27958).

The entire HCV genome is also disclosed, for example, under Genbank accession nos. AAB67038; NP671491; AAB67036; AAG02099; AAB67037; and AAP69952.

An example of a human CD81 is disclosed under Genbank accession no. NM004356.

A “polynucleotide”, “nucleic acid” or “nucleic acid molecule” includes the phosphate ester polymeric form of ribonucleosides (adenosine, guanosine, uridine or cytidine; “RNA molecules”) or deoxyribonucleosides (deoxyadenosine, deoxyguanosine, deoxythymidine, or deoxycytidine; “DNA molecules”), or any phosphoester analogs thereof, such as phosphorothioates and thioesters, in single stranded form, double-stranded form or otherwise.

A “polynucleotide sequence”, “nucleic acid sequence” or “nucleotide sequence” is a series of nucleotide bases (also called “nucleotides”) in a nucleic acid, such as DNA or RNA, and means any chain of two or more nucleotides.

A “coding sequence” or a sequence “encoding” an expression product, such as a RNA, polypeptide, protein, or enzyme, is a nucleotide sequence that, when expressed, results in production of the product.

The term “gene” means a DNA sequence that codes for or corresponds to a particular sequence of ribonucleotides or amino acids which comprise all or part of one or more RNA molecules, proteins or enzymes, and may or may not include regulatory DNA sequences, such as promoter sequences, which determine, for example, the conditions under which the gene is expressed. Genes may be transcribed from DNA to RNA which may or may not be translated into an amino acid sequence.

The present invention includes any nucleic acid fragment encoding a polypeptide taken from of any of SEQ ID NOs: 1 and 2. An embodiment of the invention includes any nucleic acid including at least about 24 (e.g., 25, 26, 27, 28, 29, 30, 31, 32, 33, 34), preferably at least about 35 (e.g., 25, 26, 27, 28, 29, 30, 31, 32, 33 or 34), more preferably at least about 45 (e.g., 35, 36, 37, 38, 39, 40, 41, 42, 43 or 44), and most preferably at least about 126 or more contiguous nucleotides (e.g., 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 1000 or 1200) from any polynucleotide encoding a polypeptide of any of SEQ ID NOs: 1 or 2 (e.g., any of SEQ ID NOs: 3-139).

An embodiment of the invention includes any polynucleotide that encodes any of the polypeptides of the invention (e.g., any of SEQ ID NOs: 3-139), particularly any of SEQ ID NOs: 3, 7, 9, 10, 12-28, 30-38, 40, 42, 44-60, 62-68, 70, 71, 73-103, 106-120, 121-130, 132-138.

As used herein, the term “oligonucleotide” refers to a nucleic acid, generally of no more than about 100 nucleotides (e.g., 30, 40, 50, 60, 70, 80, or 90), that may be hybridizable to a genomic DNA molecule, a cDNA molecule, or an mRNA molecule encoding a gene, mRNA, cDNA, or other nucleic acid of interest. Oligonucleotides can be labeled, e.g., by incorporation of 32P-nucleotides, 3H-nucleotides, 14C-nucleotides, 35S-nucleotides or nucleotides to which a label, such as biotin, has been covalently conjugated. In one embodiment, a labeled oligonucleotide can be used as a probe to detect the presence of a nucleic acid. In another embodiment, oligonucleotides (one or both of which may be labeled) can be used as PCR primers, either for cloning full length or a fragment of the gene, or to detect the presence of nucleic acids. Generally, oligonucleotides are prepared synthetically, preferably on a nucleic acid synthesizer.

A “protein sequence”, “peptide sequence” or “polypeptide sequence” or “amino acid sequence” includes a series of two or more amino acids in a protein, peptide or polypeptide.

“Protein”, “peptide” or “polypeptide” includes a contiguous string of two or more amino acids. An embodiment of the invention includes any polypeptide comprising at least about 7 (e.g., 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 or 19), more preferably at least about 20 (e.g., 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40), and yet more preferably at least about 42 (e.g., 43, 44, 45, 46, 47, 48, 49, 50, 60, 70, 80, 90, 100, 110, 120 or 130) or more contiguous amino acid residues from SEQ ID NOs: 1 or 2 (e.g., any of SEQ ID NOs: 3-139).

The scope of the present invention includes any of the polypeptides described herein (e.g., any of SEQ ID NOs: 3-139) comprising either D- or L-amino acids or one or more of either. For example, the present invention includes L-Val-L-Ser-L-Phe-L-Ala-L-Ile-L-Lys-L-Trp-L-Glu-L-Tyr-L-Val-L-Leu-L-Leu-L-Leu-L-Phe-L-Leu-L-Leu (SEQ ID NO: 77) as well as, for example, D-Val-D-Ser-D-Phe-D-Ala-D-Ile-D-Lys-D-Trp-D-Glu-D-Tyr-D-Val-D-Leu-D-Leu-D-Leu-D-Phe-D-Leu-D-Leu and L-Val-D-Ser-L-Phe-L-Ala-D-Ile-L-Lys-L-Trp-D-Glu-L-Tyr-L-Val-L-Leu-D-Leu-L-Leu-L-Phe-L-Leu-D-Leu. The present invention also includes any of the polypeptides described herein along with the reverse version of each peptide. For example, the present invention includes V-S-F-A-I-K-W-E-Y-V-L-L-L-F-L-L (SEQ ID NO: 77) along with the reverse of this peptide: L-L-F-L-L-L-V-Y-E-W-K-I-A-F-S-V.

The terms “isolated polynucleotide” or “isolated polypeptide” include a polynucleotide (e.g., RNA or DNA molecule, or a mixed polymer) or a polypeptide, respectively, which are partially or fully separated from other components that are normally found in cells or in recombinant DNA expression systems. These components include, but are not limited to, cell membranes, cell walls, ribosomes, polymerases, serum components and extraneous genomic sequences.

An isolated polynucleotide or polypeptide will, preferably, be an essentially homogeneous composition of molecules but may contain some heterogeneity.

“Amplification” of DNA as used herein includes the use of polymerase chain reaction (PCR) to increase the concentration of a particular DNA sequence within a mixture of DNA sequences. For a description of PCR see Saiki, et al., Science (1988) 239:487.

The term “host cell” includes any cell of any organism that is selected, modified, transfected, transformed, grown, or used or manipulated in any way, for the production of a substance by the cell, for example the expression or replication, by the cell, of a gene, a DNA or RNA sequence or a protein (e.g., chinese hamster ovary (CHO) cells and bacterial cells including E. coli (e.g., BL21, BL21DE3, DH5, DH5a and HB101 cells)).

The nucleotide sequence of a nucleic acid may be determined by any method known in the art (e.g., chemical sequencing or enzymatic sequencing). “Chemical sequencing” of DNA includes methods such as that of Maxam and Gilbert (1977) (Proc. Natl. Acad. Sci. USA 74:560), in which DNA is randomly cleaved using individual base-specific reactions. “Enzymatic sequencing” of DNA includes methods such as that of Sanger (Sanger, et al., (1977) Proc. Natl. Acad. Sci. USA 74:5463).

The nucleic acids herein may be flanked by natural regulatory (expression control) sequences, or may be associated with heterologous sequences, including promoters, internal ribosome entry sites (IRES) and other ribosome binding site sequences, enhancers, response elements, suppressors, signal sequences, polyadenylation sequences, introns, 5′- and 3′-non-coding regions, and the like.

In general, a “promoter” or “promoter sequence” is a DNA regulatory region capable of binding an RNA polymerase in a cell (e.g., directly or through other promoter-bound proteins or substances) and initiating transcription of a coding sequence. A promoter sequence is, in general, bounded at its 3′ terminus by the transcription initiation site and extends upstream (5′ direction) to include the minimum number of bases or elements necessary to initiate transcription at any level. Within the promoter sequence may be found a transcription initiation site (conveniently defined, for example, by mapping with nuclease S1), as well as protein binding domains (consensus sequences) responsible for the binding of RNA polymerase. The promoter may be operably associated with other expression control sequences, including enhancer and repressor sequences or with a nucleic acid of the invention. Promoters which may be used to control gene expression include, but are not limited to, cytomegalovirus (CMV) promoter (U.S. Pat. Nos. 5,385,839 and 5,168,062), the SV40 early promoter region (Benoist, et al., (1981) Nature 290:304-310), the promoter contained in the 3′ long terminal repeat of Rous sarcoma virus (Yamamoto, et al., (1980) Cell 22:787-797), the herpes thymidine kinase promoter (Wagner, et al., (1981) Proc. Natl. Acad. Sci. USA 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster, et al., (1982) Nature 296:39-42); prokaryotic promoters such as the β-lactamase promoter (VIIIa-Komaroff, et al., (1978) Proc. Natl. Acad. Sci. USA 75:3727-3731), or the tac promoter (DeBoer, et al., (1983) Proc. Natl. Acad. Sci. USA 80:21-25); see also “Useful proteins from recombinant bacteria” in Scientific American (1980) 242:74-94; and promoter elements from yeast or other fungi such as the Gal 4 promoter, the ADH (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter or the alkaline phosphatase promoter.

A coding sequence is “under the control of”, “functionally associated with” or “operably associated with” a transcriptional and translational control sequence in a cell (e.g., a promoter), for example, when the sequence directs RNA polymerase mediated transcription of the coding sequence into RNA, preferably mRNA, which then may be RNA spliced (if it contains introns) and, optionally, translated into a protein encoded by the coding sequence.

The terms “express” and “expression” mean allowing or causing the information in a gene, RNA or DNA sequence to become manifest; for example, producing a protein by activating the cellular functions involved in transcription and translation of a corresponding gene. A DNA sequence is expressed in or by a cell to form an “expression product” such as an RNA (e.g., mRNA) or a protein. The expression product itself may also be said to be “expressed” by the cell.

The term “transformation” means the introduction of a nucleic acid into a cell. The introduced gene or sequence may be called a “clone”. A host cell that receives the introduced DNA or RNA has been “transformed” and is a “transformant” or a “clone.” The DNA or RNA introduced to a host cell can come from any source, including cells of the same genus or species as the host cell, or from cells of a different genus or species. The introduced nucleic acid can be maintained in the cell episomally or may be integrated into a chromosome of the cell.

The term “vector” includes a vehicle (e.g., a plasmid) by which a DNA or RNA sequence can be introduced into a host cell, so as to transform the host and, optionally, promote expression and/or replication of the introduced sequence.

Vectors that can be used in this invention include plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles that may facilitate introduction of the nucleic acids into the genome of the host. Plasmids are the most commonly used form of vector but all other forms of vectors which serve a similar function and which are, or become, known in the art are suitable for use herein. See, e.g., Pouwels, et al., Cloning Vectors: A Laboratory Manual, 1985 and Supplements, Elsevier, N.Y., and Rodriguez et al. (eds.), Vectors: A Survey of Molecular Cloning Vectors and Their Uses, 1988, Buttersworth, Boston, Mass.

The term “expression system” means a host cell and compatible vector which, under suitable conditions, can express a protein or nucleic acid which is carried by the vector and introduced to the host cell. Common expression systems include E. coli host cells and plasmid vectors, insect host cells and Baculovirus vectors, and mammalian host cells and vectors.

Expression of nucleic acids encoding the E1-E2 polypeptides of this invention (e.g., SEQ ID NOs: 3-139) can be carried out by conventional methods in either prokaryotic or eukaryotic cells. Although E. coli host cells are employed most frequently in prokaryotic systems, many other bacteria, such as various strains of Pseudomonas and Bacillus, are known in the art and can be used as well. Suitable host cells for expressing nucleic acids encoding the E1-E2 polypeptides include prokaryotes and higher eukaryotes. Prokaryotes include both gram-negative and gram-positive organisms, e.g., E. coli and B. subtilis. Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents.

Prokaryotic host-vector systems include a wide variety of vectors for many different species. A representative vector for amplifying DNA is pBR322 or many of its derivatives (e.g., pUC18 or 19). Vectors that can be used to express the E1-E2 polypeptides (e.g., SEQ ID NOs: 3-139) include, but are not limited to, those containing the lac promoter (pUC-series); trp promoter (pBR322-trp); lpp promoter (the pIN-series); lambda-pP or pR promoters (pOTS); or hybrid promoters such as ptac (pDR540). See Brosius et al., “Expression Vectors Employing Lambda-, trp-, lac-, and Ipp-derived Promoters”, in Rodriguez and Denhardt (eds.) Vectors: A Surve of Molecular Clonin Vectors and Their Uses, 1988, Buttersworth, Boston, pp. 205-236. Many polypeptides can be expressed, at high levels, in an E. coli/T7 bacteriophage expression system as disclosed in U.S. Pat. Nos. 4,952,496, 5,693,489 and 5,869,320 and in Davanloo, P., et al., (1984) Proc. Natl. Acad. Sci. USA 81: 2035-2039; Studier, F. W., et al., (1986) J. Mol. Biol. 189: 113-130; Rosenberg, A. H., et al., (1987) Gene 56: 125-135; and Dunn, J. J., et al., (1988) Gene 68: 259.

Higher eukaryotic tissue culture cells may also be used for the recombinant production of the E1-E2 polypeptides of the invention (e.g., SEQ ID NOs: 3-139). Although any higher eukaryotic tissue culture cell line might be used, including insect baculovirus expression systems, mammalian cells are preferred. Transformation or transfection and propagation of such cells have become a routine procedure. Examples of useful cell lines include HeLa cells, chinese hamster ovary (CHO) cell lines, J774 cells, Caco2 cells, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines. Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site. RNA splice sites (if genomic DNA is used), a polyadenylation site, and/or a transcription termination site. These vectors also, usually, contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as adenovirus, SV40, parvoviruses, vaccinia virus, or cytomegalovirus. Examples of expression vectors include pCR®3.1, pcDNA1, pCD (Okayama, et al., (1985) Mol. Cell. Biol. 5:1136), pMC1neo Poly-A (Thomas, et al., (1987) Cell 51:503), pREP8, pSVSPORT and derivatives thereof, and baculovirus vectors such as pAC373 or pAC610.

The present invention also includes fusions which include any of the E1-E2 polypeptides (e.g., SEQ ID NOs: 3-139) and E1-E2 polynucleotides encoding said polypeptides of the present invention or a fragment thereof (discussed above) and a second polypeptide or polynucleotide moiety, which may be referred to as a “tag”. The fused polypeptides of the invention may be conveniently constructed, for example, by insertion of a polynucleotide of the invention or fragment thereof into an expression vector. The fusions of the invention may include tags which facilitate purification or detection. Such tags include glutathione-S-transferase (GST), hexahistidine (His6) tags, maltose binding protein (MBP) tags, haemagglutinin (HA) tags, cellulose binding protein (CBP) tags and myc tags. Detectable tags such as 32P, 35S, 3H, 99mTc, 123I, 111In, 68Ga, 18F, 125I, 131I, 113mIn, 76Br, 67Ga, 99mTc, 123I, 111In, and 68Ga may also be used to label the polypeptides and polynucleotides of the invention. Methods for constructing and using such fusions are very conventional and well known in the art.

Modifications (e.g., post-translational modifications) that occur in a polypeptide often will be a function of how it is made. For polypeptides made by expressing a cloned gene in a host, for instance, the nature and extent of the modifications, in large part, will be determined by the host cell's post-translational modification capacity and the modification signals present in the polypeptide amino acid sequence. For instance, as is well known, glycosylation often does not occur in bacterial hosts such as E. coli. Accordingly, when glycosylation is desired, a polypeptide can be expressed in a glycosylating host, generally a eukaryotic cell. Insect cells often carry out post-translational glycosylations which are similar to those of mammalian cells. For this reason, insect cell expression systems have been developed to express, efficiently, proteins having glycosylation. An insect cell which may be used in this invention is any cell derived from an organism of the class Insecta. For example, the insect can be Spodoptera fruigiperda (Sf9 or Sf21) or Trichoplusia ni (High 5). Examples of insect expression systems that can be used with the present invention, for example to produce E1-E2 polypeptide (e.g., SEQ ID NOs: 3-139), include Bac-To-Bac (Invitrogen Corporation, Carlsbad, Calif.) or Gateway (Invitrogen Corporation, Carlsbad, Calif.). If desired, deglycosylation enzymes can be used to remove carbohydrates attached during production in eukaryotic expression systems.

Other modifications may also include addition of aliphatic esters or amides to the polypeptide carboxyl terminus. The present invention also includes analogs of the E1-E2 polypeptides (e.g., SEQ ID NOs: 3-139) which contain modifications, such as incorporation of unnatural amino acid residues, or phosphorylated amino acid residues such as phosphotyrosine, phosphoserine or phosphothreonine residues. Other potential modifications include sulfonation, biotinylation, or the addition of other moieties. For example, the E1-E2 polypeptides of the invention (e.g., SEQ ID NOs: 3-139) may be appended with a polymer which increases the half-life of the peptide in the body of a subject. Polymers include polyethylene glycol (PEG) (e.g., PEG with a molecular weight of 2 kDa, 5 kDa, 10 kDa, 12 kDa, 20 kDa, 30 kDa and 40 kDa), dextran and monomethoxypolyethylene glycol (mPEG).

The peptides of the invention may also be cyclized. Specifically, the amino- and carboxy-terminal residues of an E1-E2 polypeptide (e.g., SEQ ID NOs: 3-139) or two internal residues of an E1-E2 polypeptide of the invention (e.g., SEQ ID NOs: 3-139) or a terminal and an internal residue of an E1-E3 polypeptide of the invention (e.g., SEQ ID NOs: 3-139) can be fused to create a cyclized peptide. Methods for cyclizing peptides are conventional and very well known in the art; for example see Gurrath, et al., (1992) Eur. J. Biochem. 210:911-921.

The present invention contemplates any superficial or slight modification to the amino acid or nucleotide sequences which correspond to the E1-E2 polypeptides of the invention (e.g., SEQ ID NOs: 3-139). In particular, the present invention contemplates sequence conservative variants of the nucleic acids which encode the polypeptides of the invention. “Sequence-conservative variants” of a polynucleotide sequence are those in which a change of one or more nucleotides in a given codon results in no alteration in the amino acid encoded at that position. Function-conservative variants of the polypeptides of the invention (e.g., any of SEQ ID NOs: 3-139) are also contemplated by the present invention along with nucleic acids encoding the variants. “Function-conservative variants” are those in which one or more amino acid residues in a protein or enzyme have been changed without significantly altering the overall conformation and/or function of the polypeptide, including, but, by no means, limited to, replacement of an amino acid with one having similar properties. Amino acids with similar properties are well known in the art. For example, polar/hydrophilic amino acids which may be interchangeable include asparagine, glutamine, serine, cysteine, threonine, lysine, arginine, histidine, aspartic acid and glutamic acid; nonpolar/hydrophobic amino acids which may be interchangeable include glycine, alanine, valine, leucine, isoleucine, proline, tyrosine, phenylalanine, tryptophan and methionine; acidic amino acids which may be interchangeable include aspartic acid and glutamic acid and basic amino acids which may be interchangeable include histidine, lysine and arginine.

The present invention includes E1-E2 polynucleotides (e.g., any polynucleotide encoding a polypeptide comprising an amino acid sequence selected from SEQ ID NOs: 3-139) and fragments thereof as well as nucleic acids which hybridize to the polynucleotides. Preferably, the nucleic acids hybridize under low stringency conditions, more preferably under moderate stringency conditions and most preferably under high stringency conditions. A nucleic acid molecule is “hybridizable” to another nucleic acid molecule, such as a cDNA, genomic DNA, or RNA, when a single stranded form of the nucleic acid molecule can anneal to the other nucleic acid molecule under the appropriate conditions of temperature and solution ionic strength (see Sambrook, et al., supra). The conditions of temperature and ionic strength determine the “stringency” of the hybridization. Typical low stringency hybridization conditions are 55° C., 5×SSC, 0.1% SDS, 0.25% milk, and no formamide at 42° C.; or 30% formamide, 5×SSC, 0.5% SDS at 42° C. Typical, moderate stringency hybridization conditions are similar to the low stringency conditions except the hybridization is carried out in 40% formamide, with 5× or 6×SSC at 42° C. High stringency hybridization conditions are similar to low stringency conditions except the hybridization conditions are carried out in 50% formamide, 5× or 6×SSC and, optionally, at a higher temperature (e.g., higher than 42° C.: 57° C., 59° C., 60° C., 62° C., 63° C., 65° C. or 68° C.). In general, SSC is 0.15M NaCl and 0.015M Na-citrate. Hybridization requires that the two nucleic acids contain complementary sequences, although, depending on the stringency of the hybridization, mismatches between bases are possible. The appropriate stringency for hybridizing nucleic acids depends on the length of the nucleic acids and the degree of complementation, variables well known in the art. The greater the degree of similarity or homology between two nucleotide sequences, the higher the stringency under which the nucleic acids may hybridize. For hybrids of greater than 100 nucleotides in length, equations for calculating the melting temperature have been derived (see Sambrook, et al., supra, 9.50-9.51). For hybridization with shorter nucleic acids, i.e., oligonucleotides, the position of mismatches becomes more important, and the length of the oligonucleotide determines its specificity (see Sambrook, et al., supra).

Also included in the present invention are polynucleotides comprising nucleotide sequences and polypeptides comprising amino acid sequences which are at least about 70% identical, preferably at least about 80% identical, more preferably at least about 90% identical and most preferably at least about 95% identical (e.g., 95%, 96%, 97%, 98%, 99%, 100%) to the reference E1-E2 nucleotide and amino acid sequences (e.g., SEQ ID NOs: 3-139) of the invention, when the comparison is performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences. Polypeptides comprising amino acid sequences which are at least about 70% similar, preferably at least about 80% similar, more preferably at least about 90% similar and most preferably at least about 95% similar (e.g., 95%, 96%, 97%, 98%, 99%, 100%) to the reference E1-E2 amino acid sequence of any of SEQ ID NOs: 3-139, when the comparison is performed with a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences, are also included in the present invention.

Sequence identity refers to exact matches between the nucleotides or amino acids of two sequences which are being compared. Sequence similarity refers to both exact matches between the amino acids of two polypeptides which are being compared in addition to matches between nonidentical, biochemically related amino acids. Biochemically related amino acids which share similar properties and may be interchangeable are discussed above.

The following references regarding the BLAST algorithm are herein incorporated by reference: BLAST ALGORITHMS: Altschul, S. F., et al., (1990) J. Mol. Biol. 215:403-410; Gish, W., et al., (1993) Nature Genet. 3:266-272; Madden, T. L., et al., (1996) Meth. Enzymol. 266:131-141; Altschul, S. F., et al., (1997) Nucleic Acids Res. 25:3389-3402; Zhang, J., et al., (1997) Genome Res. 7:649-656; Wootton, J. C., et al., (1993) Comput. Chem. 17:149-163; Hancock, J. M., et al., (1994) Comput. Appl. Biosci. 10:67-70; ALIGNMENT SCORING SYSTEMS: Dayhoff, M. O., et al., “A model of evolutionary change in proteins.” in Atlas of Protein Sequence and Structure, (1978) vol. 5, suppl. 3. M. O. Dayhoff (ed.), pp. 345-352, Natl. Biomed. Res. Found., Washington, D.C.; Schwartz, R. M., et al., “Matrices for detecting distant relationships.” in Atlas of Protein Sequence and Structure, (1978) vol. 5, suppl. 3.” M. O. Dayhoff (ed.), pp. 353-358, Natl. Biomed. Res. Found., Washington, D.C.; Altschul, S. F., (1991) J. Mol. Biol. 219:555-565; States, D. J., et al., (1991) Methods 3:66-70; Henikoff, S., et al., (1992) Proc. Natl. Acad. Sci. USA 89:10915-10919; Altschul, S. F., et al., (1993) J. Mol. Evol. 36:290-300; ALIGNMENT STATISTICS: Karlin, S., et al., (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268; Karlin, S., et al., (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877; Dembo, A., et al., (1994) Ann. Prob. 22:2022-2039; and Altschul, S. F. “Evaluating the statistical significance of multiple distinct local alignments.” in Theoretical and Computational Methods in Genome Research (S. Suhai, ed.), (1997) pp. 1-14, Plenum, New York.

Protein Purification

The polypeptides of this invention (e.g., SEQ ID NOs: 3-139) can be purified by standard methods, including, but not limited to, salt or alcohol precipitation, affinity chromatography (e.g., used in conjunction with a purification tagged E1-E2 polypeptide as discussed above), preparative disc-gel electrophoresis, isoelectric focusing, high pressure liquid chromatography (HPLC), reversed-phase HPLC, gel filtration, cation and anion exchange and partition chromatography, and countercurrent distribution. Such purification methods are well known in the art and are disclosed, e.g., in “Guide to Protein Purification”, Methods in Enzymology, Vol. 182, M. Deutscher, Ed., 1990, Academic Press, New York, N.Y.

Where an E1-E2 polypeptide (e.g., any of SEQ ID NOs: 3-139) is being isolated from a cellular or tissue source, it is preferable to include one or more inhibitors of proteolytic enzymes in the assay system, such as phenylmethanesulfonyl fluoride (PMSF), Pefabloc SC, pepstatin, leupeptin, chymostatin and EDTA.

Pharmaceutical Compositions

The present invention includes methods for using a pharmaceutical composition comprising a peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) and a pharmaceutically acceptable carrier for treating a Flaviviridae infection. The pharmaceutical compositions may be prepared by any methods well known in the art of pharmacy; see, e.g., Gilman, et al., (eds.) (1990), The Pharmacological Bases of Therapeutics, 8th Ed., Pergamon Press; A. Gennaro (ed.), Remington's Pharmaceutical Sciences, 18th Edition, (1990), Mack Publishing Co., Easton, Pa.; Avis, et al., (eds.) (1993) Pharmaceutical Dosage Forms Parenteral Medications Dekker, New York; Lieberman, et al., (eds.) (1990) Pharmaceutical Dosage Forms: Tablets Dekker, New York; and Lieberman, et al., (eds.) (1990), Pharmaceutical Dosage Forms: Disperse Systems Dekker, New York.

A pharmaceutical composition containing an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) can be prepared using conventional pharmaceutically acceptable excipients and additives and conventional techniques. Such pharmaceutically acceptable excipients and additives include non-toxic compatible fillers, binders, disintegrants, buffers, preservatives, anti-oxidants, lubricants, flavorings, thickeners, coloring agents, emulsifiers and the like. All routes of administration are contemplated including, but not limited to, parenteral (e.g., subcutaneous, intravenous, intraperitoneal, intramuscular) and non-parenteral (e.g., oral, transdermal, intranasal, intraocular, sublingual, inhalation, rectal and topical).

Unit forms of administration include oral forms such as tablets, capsules, powders, cachets, granules and solutions or suspensions, sublingual and buccal forms of administration, aerosols, implants, subcutaneous, intramuscular, intravenous, intranasal, intraocular, subcutaneous or rectal forms of administration.

When a solid composition is prepared in the form of tablets, e.g., a wetting agent such as sodium lauryl sulfate can be added to micronized or non-micronized compounds and mixed with a pharmaceutical vehicle such as silica, gelatin starch, lactose, magnesium stearate, talc, gum arabic or the like. The tablets can be coated with sucrose, various polymers, or other appropriate substances. Tablets can be treated so as to have a prolonged or delayed activity and so as to release a predetermined amount of active principle continuously or at predetermined intervals, e.g., by using ionic resins and the like.

A preparation in the form of gelatin capsules may be obtained, e.g., by mixing an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) with a diluent, such as a glycol or a glycerol ester, and incorporating the resulting mixture into soft or hard gelatin capsules.

A preparation in the form of a syrup or elixir can contain an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139), e.g., with a sweetener, methylparaben and propylparaben as antiseptics, flavoring agents and an appropriate color.

Water-dispersible powders or granules can contain an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) mixed, e.g., with dispersants, wetting agents or suspending agents, such as polyvinylpyrrolidone, as well as with sweeteners and/or other flavoring agents.

Rectal administration may be provided by using suppositories which may be prepared, e.g., with binders melting at the rectal temperature, for example cocoa butter or polyethylene glycols.

Parenteral, intranasal or intraocular administration may be provided by using, e.g., aqueous suspensions, isotonic saline solutions or sterile and injectable solutions containing pharmacologically compatible dispersants and/or solubilizers, for example, propylene glycol or polyethylene glycol.

Thus, to prepare an aqueous solution for intravenous injection, it is possible to use a co-solvent, e.g., an alcohol such as ethanol or a glycol such as polyethylene glycol or propylene glycol, and a hydrophilic surfactant such as Tween® 80. An oily solution injectable intramuscularly can be prepared, e.g., by solubilizing the active principle with a triglyceride or a glycerol ester.

Transdermal administration can be provided by using patches in the form of a multilaminate, or with a reservoir, containing an a peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) and an appropriate solvent.

Administration by inhalation can be provided by using, e.g., an aerosol containing sorbitan trioleate or oleic acid, for example, together with trichlorofluoromethane, dichlorofluoromethane, dichlorotetrafluoroethane or any other biologically compatible propellant gas; it is also possible to use a system containing an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139), by itself or associated with an excipient, in powder form.

An E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) can also be formulated as microcapsules or microspheres, e.g., liposomes, optionally with one or more carriers or additives.

Implants are among the prolonged release forms which can be used in the case of chronic treatments. They can be prepared in the form of an oily suspension or in the form of a suspension of microspheres in an isotonic medium.

Methods of the present invention can include administration of an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with, for example, one or more known anti-viral agents. The administration and dosage of such agents is typically as according to the schedule listed in the product information sheet of the approved agents, in the Physicians' Desk Reference 2003 (Physicians' Desk Reference, 57th Ed); Medical Economics Company; ISBN: 1563634457; 57th edition (November 2002), as well as therapeutic protocols well known in the art.

Suitable anti-viral agents, with which a peptide of the invention (e.g., any of SEQ ID NOs: 3-139) can be administered or mixed or formulated include, but are not limited to, pegylated or unpegylated interferon alfa-2a, pegylated or unpegylated interferon alfa-2b, pegylated or unpegylated interferon alfa-2c, pegylated or unpegylated interferon alfa n-1, pegylated or unpegylated interferon alfa n-3 and pegylated, unpegylated consensus interferon or albumin-interferon-alpha.

The term “interferon alpha” as used herein means the family of highly homologous species-specific proteins that inhibit viral replication and cellular proliferation and modulate immune response. Typical suitable interferon-alphas include, but are not limited to, recombinant interferon alpha-2b, recombinant interferon alpha-2a, recombinant interferon alpha-2c, alpha 2 interferon, interferon alpha-n1 (INS), a purified blend of natural alpha interferons, a consensus alpha interferon such as those described in U.S. Pat. Nos. 4,897,471 and 4,695,623 (especially Examples 7, 8 or 9 thereof), or interferon alpha-n3, a mixture of natural alpha interferons.

Interferon alfa-2a is sold as ROFERON-A® by Hoffmann-La Roche (Nutley, N.J.).

Interferon alfa-2b is sold as INTRON-A® by Schering Corporation (Kenilworth, N.J.). Interferon alfa-2b is also sold, in combination with ribavirin, as REBETRON® by Schering Corporation (Kenilworth, N.J.). The manufacture of interferon alpha 2b is described, for example, in U.S. Pat. No. 4,530,901.

Interferon alfa-n3 is a mixture of natural interferons sold as ALFERON N INJECTION® by Hemispherx Biopharma, Inc. (Philadelphia, Pa.).

Interferon alfa-n1 (INS) is a mixture of natural interferons sold as WELLFERON® by Glaxo-Smith-Kline (Research Triangle Park, N.C.).

Consensus interferon is sold as INFERGEN® by Intermune, Inc. (Brisbane, Calif.).

Interferon alfa-2c is sold as BEROFOR® by Boehringer Ingelheim Pharmaceutical, Inc. (Ridgefield, Conn.).

A purified blend of natural interferons is sold as SUMIFERON® by Sumitomo; Tokyo, Japan.

A multi-subtype natural interferon derived from human white blood cells is sold by Viragen, Inc (Plantation, Fla.), as Omniferon™. Viragen, Inc. also sells a highly purified, multi-subtype, natural human alpha interferon derived from human white blood cells, as Multiferon™. (U.S. Pat. No. 6,743,624).

Pegylated interferon alpha may also be administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139). The term “pegylated interferon alpha” as used herein means polyethylene glycol modified conjugates of interferon alpha, preferably interferon alpha-2a and alpha-2b. The preferred polyethylene-glycol-interferon alpha-2b conjugate is PEG 12000-interferon alpha-2b. The phrases “12,000 molecular weight polyethylene glycol conjugated interferon alpha” and “PEG 12000-IFN alpha” as used herein include conjugates such as are prepared according to the methods of International Application No. WO 95/13090 and containing urethane linkages between the interferon alpha-2a or -2b amino groups and polyethylene glycol having an average molecular weight of 12000. The pegylated inteferon alpha, PEG 12000-IFN-alpha-2b is available from Schering-Plough Research Institute, Kenilworth, N.J.

The preferred PEG 12000-interferon alpha-2b can be prepared by attaching a PEG polymer to the epsilon amino group of a lysine residue in the interferon alpha-2b molecule. A single PEG 12000 molecule can be conjugated to free amino groups on an IFN alpha-2b molecule via a urethane linkage. This conjugate is characterized by the molecular weight of PEG 12000 attached. The PEG 12000-IFN alpha-2b conjugate can be formulated as a lyophilized powder for injection.

Pegylated interferon alfa-2b is sold as PEG-INTRON® by Schering Corporation (Kenilworth, N.J.).

Pegylated interferon-alfa-2a is sold as PEGASYS® by Hoffmann-La Roche (Nutley, N.J.).

Other interferon alpha conjugates can be prepared by coupling an interferon alpha to a water-soluble polymer. A non-limiting list of such polymers include other polyalkylene oxide homopolymers such as polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof. As an alternative to polyalkylene oxide-based polymers, effectively non-antigenic materials such as dextran, polyvinylpyrrolidones, polyacrylamides, polyvinyl alcohols, carbohydrate-based polymers and the like can be used. Such interferon alpha-polymer conjugates are described, for example, in U.S. Pat. No. 4,766,106, U.S. Pat. No. 4,917,888, European Patent Application No. 0 236 987 or 0 593 868 or International Publication No. WO 95/13090.

Pharmaceutical compositions of pegylated interferon alpha suitable for parenteral administration can be formulated with a suitable buffer, e.g., Tris-HCl, acetate or phosphate such as dibasic sodium phosphate/monobasic sodium phosphate buffer, and pharmaceutically acceptable excipients (e.g., sucrose), carriers (e.g. human plasma albumin), toxicity agents (e.g., NaCl), preservatives (e.g., thimerosol, cresol or benzyl alcohol), and surfactants (e.g., tween or polysorbates) in sterile water for injection. The pegylated interferon alpha can be stored as lyophilized powders under refrigeration at 2°-8° C. The reconstituted aqueous solutions are stable when stored between 2° and 8° C. and used within 24 hours of reconstitution. See for example U.S. Pat. Nos. 4,492,537; 5,762,923 and 5,766,582. The reconstituted aqueous solutions may also be stored in pre-filled, multi-dose syringes such as those useful for delivery of drugs such as insulin. Typical, suitable syringes include systems comprising a prefilled vial attached to a pen-type syringe such as the NOVOLET® Novo Pen available from Novo Nordisk or the REDIPEN®, available from Schering Corporation, Kenilworth, N.J. Other syringe systems include a pen-type syringe comprising a glass cartridge containing a diluent and lyophilized pegylated interferon alpha powder in a separate compartment.

In an embodiment of the invention, one or more other anti-viral substances may be administered with one or more E1-E2 peptides of the present invention (e.g., any of SEQ ID NOs: 3-139).

Other types of compounds which may be administered with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) include, but are not limited to, ribonucleoside analogues, IMPDH inhibitors, N-glycosylation inhibitors, N3 protease inhibitors, NS5B inhibitors, immunomodulatory compounds and CTP synthase inhibitors, thiazolidine derivatives, benzanilides, phenanthrenequinones, helicase inhibitors, polymerase inhibitors, antisense phosphothioate oligodeoxynucleotides, IRES-dependent translation inhibitors, nuclease resistant ribozymes, 1-amino-alkyloyclohexanes, alkyl lipids, antioxidants, squalene, amantadine, bile acids, N-(phosphonoacetyl)-L-aspartic acid, benzenedicarboxamides, polyadenylic acid derivatives, 2′,3′ dideoxyinosine and benzimidazoles.

In an embodiment of the present invention, ribavirin

is administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139). Ribavirin is sold as REBETOL® by Schering Corporation; Kenilworth, N.J. Its manufacture and formulation is described, for example, in U.S. Pat. No. 4,211,771. A combination of ribavirin and recombinant interferon alfa-2b (REBETRON®; Schering Corporation; Kenilworth, N.J.) may also be administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

In another embodiment of the invention, gemcitabine

is administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139). Gemcitabine is sold as GEMZAR® by Eli Lilly and Co. (Indianapolis, Ind.).

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with VX497

An embodiment of the invention comprises administering mycophenolate mofetil (MMF; 2-morpholinoethyl (E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate) in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139). MMF is sold as CellCept® by Roche Laboratories; Nutley, N.J.

Another embodiment comprises administerina EICAR

in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

An embodiment of the present invention comprises administering tiazofurin

in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

Another embodiment of the invention comprises administering deoxynojirimycin and/or derivatives thereof, such as N-nonyl-deoxynojirimycin (De Clercq et al., Mini Rev Med. Chem. 2(2):163-75 (2002)) or n-butyl deoxynojirimycin (nB-DNJ; Ouzounov et al., Antiviral Res. 55(3):425-35 (2002)), in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

Another embodiment of the invention comprises administering colchicine

in association with an E1-E2 peptide of the invention (e.g., any of SEQ ID NOs: 3-139).

In one embodiment, an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with albumin-interferon alpha (ALBUFERON™). Albumin-interferon alpha is interferon-α fused to human serum albumin. ALBUFERON™ is available from Human Genome Sciences, Rockville, Md. ALBUFERON™ has been shown to be effective for treatment of hepatitis C virus infections (Blaire et al., J. Pharm and Exp. Therap. 303(2): 540-548 (2002)).

In another embodiment, BILN-2061

is administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

In another embodiment, thymalfasin (e.g., ZADAXIN™) is administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139). ZADAXIN™ is available from SciClone Pharmaceuticals International, Ltd., San Mateo, Calif.

In yet another embodiment, isatoribine

is administered in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

In another embodiment, an E1-E2 peptide of the invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with an NS5B inhibitor such as NM283 or NM107 (Idenix Pharmaceuticals; Cambridge, Mass.).

In another embodiment, an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with SCH68631

or SCH351633

In a further embodiment, an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with any of the P1 variants of Elgin c disclosed in Qasim et al., Biochemistry 36: 1598-1607 (1997).

In yet another embodiment, an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with gliotoxin

Other embodiments of the invention include administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association

with RD3-4082

or with RD3-4078

or any other protease inhibitor disclosed in Sudo et al.

A further embodiment of the invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with

Kakiuchi et al., FEBS Letters 421: 217-220 (1998)) or any other proteinase inhibitor disclosed in Kakiuchi et al.

A further embodiment of the invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with helioxanthin

A further embodiment of the invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with HE2000

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with RD4-6205

or any other protease inhibitor disclosed in Sudo et al.

An embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with cerulenin

or any other HCV RNA-dependent RNA polymerase (RdRp) inhibitor disclosed in Lohmann et al.

An embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with ceplene

An additional embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with

(DiMarco et al., J. Biol. Chem., Vol. 275 (10): 7152-71572000);

(Barbato et al., EMBO J. 19: 1195-1206 (2000)) or any of the NS3/4a protease inhibitors disclosed by Ingallinella et al., Biochemistry, 41(17): 5483-5492 (2002).

Yet another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with amantadine

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with IDN-6556

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with beta-Alethine (beta-alanyl-cysteamine disulfide).

Yet another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with naphthoquinone, 2-methylnaphthoquinone, 2-hydroxynaphthoquinone, 5-hydroxynaphthoquinone, 5,8-dihydroxynaphthoquinone, alkannin or shikonin (Takeshita et al., Analytical Biochem. 247: 242-246 (1997)).

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with 1-amino-1,3,5-trimethylcyclohexane, 1-amino-1(trans),3(trans),5-trimethylcyclohexane, 1-amino-1(cis),3(cis),5-trimethylcyclohexane, 1-amino-1,3,3,5-tetramethylcyclohexane, 1-amino-1,3,3,5,5-pentamethylcyclohexane, 1-amino-1,3,5,5-tetramethyl-3-ethylcyclohexane, 1-amino-1,5,5-trimethyl-3,3-diethylcyclohexane, 1-amino-1,5,5-trimethyl-cis-3-ethylcyclohexane, 1-amino-(1S,5S)cis-3-ethyl-1,5,5-trimethylcyclohexane, 1-amino-1,5,5-trimethyl-trans-3-ethylcyclohexane, 1-amino-(1R,5S)trans-3-ethyl-1,5,5-trimethylcyclohexane, 1-amino-1-ethyl-3,3,5,5-tetramethylcyclohexane, 1-amino-1-propyl-3,3,5,5-tetramethylcyclohexane, N-methyl-1-amino-1,3,3,5,5-pentamethylcyclohexane, N-ethyl-1-amino-1,3,3,5,5-pentamethylcyclohexane, or N-(1,3,3,5,5-pentamethylcyclohexyl) pyrrolidine or any other 1-aminoalkylcyclohexane N-methyl-D-aspartate (NMDA) inhibitors disclosed in U.S. Pat. No. 6,034,134.

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with d-α-tocopherol or any other anti-HCV compound disclosed in U.S. Pat. No. 5,922,757.

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with tauroursodeoxycholic acid, chenodeoxycholic acid, ursodeoxycholic acid

or free bile acid or any other bile acid HCV inhibitor disclosed in U.S. Pat. No. 5,846,964.

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with interleukin 23 (IL-23) or IL-27 (Matsui et al., J. Virol. 78(17):9093-104 (2004)).

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with 1,1′-[1,4-phenylenebis (methylene)]bis(4,4′-trans-(4,5,6,7,8,9-hexahydro) benzimidazoyl)piperidine, 1,1′-[1,4-phenylenebis(methylene)]bis(4,4′-benzimidazoyl) piperidine or any other anti-HCV compound disclosed in U.S. Pat. No. 5,830,905.

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with N,N′-4-[(2-benzimidazole)phenyl]-1,4-butenedicarboxamide, N,N′-4-[(2-benzimidazole)phenyl]-1,6-hexanedicarboxamide, N,N′-4-[(2-benzimidazole)phenyl]-1,8-octanedicarboxamide, N,N′-4-[(2-benzimidazole)phenyl]-1,9-nonanedicarboxamide, N,N′-4-[(2-benzimidazole)phenyl]-1,10-decanedicarboxamide or N,N′-4-[(2-benzimidazole)phenyl]-1,4-butenedicarboxamide or any other carboxamide HCV inhibitor disclosed in U.S. Pat. No. 5,633,388.

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with any of the polyadenylic acid (5′) derivatives disclosed in U.S. Pat. No. 5,496,546.

A further embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with 2′,3′-dideoxyinosine (U.S. Pat. No. 5,026,687).

An embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with

or any other benzimidazole disclosed in U.S. Pat. No. 5,891,874.

An additional embodiment of the invention comprises administering VX-950

in association with an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139).

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with viramidine

or levovirin

A further embodiment of the invention includes administering an E1-E2 peptide of the invention (e.g., any of SEQ ID NOs: 3-139) in association with IDB-1016 (4H-1-Benzopyran-4-one, 2-(2,3-dihydro-3-(4-hydroxy-3-methoxyphenyl)-2-(hydroxymethyl)-1,4-benzodioxin-6-yl)-2,3-dihydro-3,5,7-trihydroxy-, (2R-(2alpha,3beta,6(2R*,3R*)))-, mixt. with soya phosphatidylcholines; CAS No. 134499-06-2).

Another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with the anti-sense polynucleotide ISIS-14803 (d(P-thio)(G-T-G-m5C-T-m5C-A-T-G-G-T-G-m5C-A-m5C-G-G-T-m5C-T) DNA).

Yet another embodiment of the present invention comprises administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) in association with any of the E2/CD81 binding inhibitors disclosed in VanCompernolle et al. (Virology 314: 371-380 (2003)), particularly compound 5 or 7 therein.

Moreover, in an embodiment of the invention, an E1-E2 peptide of the invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with an anti-CD81 antibody (e.g., an anti-human CD81 antibody). Anti-CD81 antibodies are available commercially from several sources and can be made by any practitioner of ordinary skill in the art using conventionally known techniques. For example, BD Biosciences (San Jose, Calif.) sells an anti-human CD81 antibody (see also Oren, et al., Mol. Cell. Biol. 10: 4007 (1990); Bradbury, et al., J. Immunol. 149: 2841 (1992) or Schick, et al., J. Immunol. 151:4090 (1993)). The present invention includes embodiments wherein an E1-E2 peptide of the invention (e.g., any of SEQ ID NOs: 3-139) is administered in association with an anti-human CD81 antibody or a fragment thereof, preferably an antigen-binding fragment, thereof; including monoclonal antibodies, polyclonal antibodies, bispecific antibodies, Fab antibody fragments, F(ab)2 antibody fragments, Fv antibody fragments (e.g., VH or VL), single chain Fv antibody fragments and dsFv antibody fragments. Furthermore, the antibodies of the invention may be fully human antibodies or chimeric antibodies. Preferably, the anti-CD81 antibody is a monoclonal, fully human antibody.

Combinations of the invention include an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) “in association” with one or more (e.g., 2, 3, 4, 5) additional anti-viral agents (e.g., ribavirin, interferon alfa-2a or 2b, or pegylated interferon alfa-2a or 2b). The term “in association” indicates that the components of the combinations of the invention can be formulated into a single composition for simultaneous delivery or formulated separately into two or more compositions (e.g., a kit). Furthermore, each component of a combination of the invention can be administered to a subject at a different time than when the other component is administered; for example, each administration may be given non-simultaneously (e.g., separately or sequentially) at several intervals over a given period of time. Moreover, the separate components may be administered to a subject by the same or by a different route (e.g., orally, intravenously, subcutaneously).

The present invention further comprises compositions comprising an E1-E2 peptide of the present invention (e.g., SEQ ID NOs: 3-139) in combination or in association with one or more anti-viral agents discussed above (e.g., pegylated interferon alfa-2a or 2b or ribavirin) along with pharmaceutical compositions thereof.

In addition, the present invention comprises compositions and methods for treating HCV infection in hosts that are co-infected with HIV. Accordingly, the present invention comprises compositions (e.g., kits) comprising an E1-E2 peptide of the invention (e.g., any of SEQ ID NO: 3-139) in association with one or more HIV protease inhibitors, various nucleoside analogs, nonnucleoside reverse transcriptase inhibitors, antivirals, immunomodulators, antiinfectives, tat antagonists, and glycosidase inhibitors. Numerous examples of such anti-HIV agents are set forth in U.S. Pat. Nos. 6,100,277 and 6,245,806, both incorporated herein by reference, and include, but are not limited to, Ro 31-859, KNI-272, AZT, DDI, DDC, 3TC, D4T, PMEA, Ro 5-3335, Ro 24-7429, indinavir, ritonavir, saquinavir, nelfinavir, amprenavir, abacavir, castanospremine, castanospermine 6-butryl ester, N-butyl-1-deoxy-nojirimycin, N-butyl-1-deoxynojirimycin per-butryl ester, 097, acemannan, acyclovir, AD-439, AD-519, adefovir clipivoxil, AL-721, alpha interferon, ansamycin, beta-fluoro-ddA, BMS-232623, BMS-234475, CI-1012, cidofovir, delaviridine, EL-10, efaviren, famciclovir, FTC, hypericin, Compound Q, ISIS 2922, lobucavir, nevirapine, novapren, peptide T, octapeptide, PNU-140690, probacol, stavudine, valaciclovir, virazole, zalcitabine, ABT-378, bropirimine, gamma interferon, interleukin-2, TNF, etanercept, infliximab, fluconalzole, piritrexim, trimetrexate, daunorubicin, leukotriene B4 receptor antagonist, and analogs and prodrugs thereof. Methods for treating HCV in a patient co-infected with HIV comprising administering an E1-E2 peptide of the invention (e.g., any of SEQ ID NOs: 3-139) in association with an anti-HIV agent (e.g., as discussed herein) are also part of the present invention.

Kits

Kits of the present invention include an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139), preferably combined with a pharmaceutically acceptable carrier, in a pharmaceutical formulation, more preferably in a pharmaceutical dosage form such as a pill, a powder, an injectable liquid, a tablet, dispersible granules, a capsule, a cachet or a suppository. See for example, Gilman et al. (eds.) (1990), The Pharmacological Bases of Therapeutics, 8th Ed., Pergamon Press; and Remington's Pharmaceutical Sciences, supra, Easton, Pa.; Avis et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Medications Dekker, New York; Lieberman et al. (eds.) (1990) Pharmaceutical Dosage Forms: Tablets Dekker, New York; and Lieberman et al. (eds.) (1990), Pharmaceutical Dosage Forms: Disperse Systems Dekker, New York. In an embodiment of the invention, a kit of the invention also includes one or more other anti-viral agents, for example, any of the substances discussed above (e.g., ribavirin, pegylated interferon alfa-2a, pegylated interferon alfa-2b or an anti-HIV agent).

A kit of the invention can also include a package insert. The package insert may include information concerning the pharmaceutical compositions and dosage forms in the kit. Generally, such information aids patients and physicians in using the enclosed pharmaceutical compositions and dosage forms effectively and safely. For example, the following information regarding an E1-E2 peptide of the invention or other anti-viral agent, included in the kit, may be supplied in the insert: pharmacokinetics, pharmacodynamics, clinical studies, efficacy parameters, indications and usage, contraindications, warnings, precautions, adverse reactions, overdosage, proper dosage and administration, how supplied, proper storage conditions, references and patent information.

The E1-E2 peptide of the invention (e.g., SEQ ID NOs: 3-139) and the other anti-viral agent can be supplied, in the kit, as separate compositions or combined into a single composition. For example, the compositions can be supplied within a single, common pharmaceutical dosage form (e.g., pill or tablet) as in separate pharmaceutical dosage forms (e.g., two separate pills or tablets).

Dosage and Administration

Pharmaceutical composition of the invention may be administered, for example, by any parenteral (e.g., subcutaneous injection, intramuscular injection, intravenous injection) or non-parenteral route (e.g., orally, nasally).

Pills and capsules of the invention can be administered orally. Injectable compositions can be administered with medical devices known in the art; for example, by injection with a hypodermic needle including the REDIPEN® or the NOVOLET® Novo Pen discussed above.

Injectable pharmaceutical compositions of the invention can also be administered with a needleless hypodermic injection device; such as the devices disclosed in U.S. Pat. No. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824 or 4,596,556.

Peptides of the invention can be administered, for example, three times a day, twice a day, once a day, three times weekly, twice weekly or once weekly.

The daily dose of an anti-viral agent administered in association with a peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) is, where possible, administered accordance with the Physicians' Desk Reference 2003 (Physicians' Desk Reference, 57th Ed); Medical Economics Company; ISBN: 1563634457; 57th edition (November 2002). The proper dosage can, however, be altered by a clinician to compensate for particular characteristics of the subject receiving the therapy depending, for example, on the potency of the compound administered, side-effects, age, weight, medical condition, overall health and response.

The term “therapeutically effective amount” means that amount of a therapeutic agent or substance (e.g., peptide of the present invention (e.g., any of SEQ ID NOs: 3-139), interferon or ribavirin) that will elicit a biological or medical response of a tissue, system, subject or host that is being sought by the administrator (such as a researcher, doctor or veterinarian) which includes alleviation of the symptoms of Flaviviridae virus (e.g., HCV) infection and the prevention, slowing or halting of progression of Flaviviridae virus (e.g., HCV) infection and its symptom(s) to any degree including prevention of the infection of a host with a Flaviviridae virus (e.g., HCV) following transplant of a liver into said host. For example, in one embodiment, a “therapeutically effective dosage” of an E1-E2 peptide of the present invention (e.g., SEQ ID NOs: 3-139) or a combination including another anti-viral agent (e.g., ribavirin and/or pegylated or unpegylated interferon alfa-2a or 2b) results in the eradication of detectable Flaviviridae Viral RNA (e.g., HCV RNA) for any period of time, for example, 12 or more weeks (e.g., 24 weeks). Detection of viral RNA in a host can be done easily using conventional, well-known methods in the art. See also the Physicians' Desk Reference (“PDR”) for the therapeutically effective dose and dosage regimens approved by the U.S. Food and Drug Administration.

A person suffering from chronic hepatitis C infection may exhibit one or more of the following signs or symptoms:

(a) elevated ALT,
(b) positive test for anti-HCV antibodies,
(c) presence of HCV as demonstrated by a positive test for HCV-RNA,
(d) clinical stigmata of chronic liver disease,
(e) hepatocelluar damage.

Ideally, though not necessarily, an infected host who is administered a composition of the invention will, eventually, exhibit no detectable HCV RNA is his body for a period of time (e.g., 12 or more weeks).

The term “no detectable HCV-RNA” in the context of the present invention means that there is less than about 100 copies of HCV-RNA per ml of serum of the patient as measured by quantitative, multi-cycle reverse transcriptase PCR (rtPCT) methodology. Such PCR based assays are conventional and very well known in the art. In general, rtPCR is performed by isolating the RNA from a specimen, reverse-transcribing it to generate cDNAs, amplifying specific nucleic acid sequences by PCR, and then using a variety of methods to detect the amplified sequences (Urdea et al., Clin. Chem. 43:1507-1511 (1997)). Preferably, a patient with no detectable HCV-RNA comprises 0 copies per ml of serum.

In one embodiment, a host administered a composition of the present invention exhibits a sustained virologic response. The term “sustained virologic response” as used in the context of the present invention means that there is no detectable HCV-RNA in the serum of patients treated in accordance with the present invention for at least 24 weeks after the end of the combined therapy treatment. Preferably, the period of sustained virologic response is at least one year—or longer—after the end of treatment.

A “therapeutically effective” dosage or amount of an E1-E2 peptide of the present invention (e.g., SEQ ID NOs: 3-139; e.g., for treating or preventing Flaviviridae virus (e.g., HCV) infection) is about 100 μg three times per day or about 300 μg once a day. However, as discussed above, this dosage may be adjusted up or down by a clinician depending on the circumstances of each particular case.

As discussed herein, methods of the present invention include administering an E1-E2 peptide of the present invention (e.g., any of SEQ ID NOs: 3-139) along with pegylated or unpegylated interferon alfa-2a, pegylated or unpegylated interferon alfa-2b, pegylated or unpegylated interferon alfa-2c, pegylated or unpegylated interferon alfa n-1, pegylated or unpegylated interferon alfa n-3, unpegylated pegylated consensus interferon, ribavirin or any combination thereof.

A therapeutically effective dosage of interferon alfa-2b (e.g., INTRON-A®), particularly for the treatment of chronic hepatitis C is 3 million IU (international units) three times a week (TIW) administered subcutaneously or intramuscularly. In patients tolerating therapy with normalization of serum alanine aminotransferase (ALT) at 16 weeks of treatment, INTRON A® therapy should be extended to 18 to 24 months (72 to 96 weeks) at 3 million IU TIW to improve the sustained response rate.

If severe adverse reactions develop during INTRON A® treatment, the dose should be modified (50% reduction) or therapy should be discontinued as indicated below. If intolerance persists after dose adjustment, INTRON A® therapy should be discontinued.

The recommended dose of PEG-interferon alfa-2b (e.g., PEG-INTRON®) regimen is from about 0.5 to about 1.5 μg/kg/week, preferably 1.0 μg/kg/week for one year.

A therapeutically effective dosage of interferon alfa-2a (e.g., ROFERON-A®), particularly for the treatment of chronic hepatitis C, is 3 MIU three times a week (TIW) administered subcutaneously or intramuscularly for 12 months (48 to 52 weeks). As an alternative, patients may be treated with an induction dose of 6 MIU TIW for the first 3 months (12 weeks) followed by 3 MIU TIW for 9 months (36 weeks).

Patients who tolerate and partially or completely respond to therapy with ROFERON-A® but relapse following its discontinuation may be re-treated. Re-treatment with either 3 MIU TIW or with 6 MIU TIW for 6 to 12 months may be considered.

A temporary dose reduction by 50% is recommended in patients who do not tolerate the prescribed dose of ROFERON-A®. If adverse events resolve, treatment with the original prescribed dose can be re-initiated. In patients who cannot tolerate the reduced dose, cessation of therapy, at least temporarily, is recommended.

The recommended dose of PEG-interferon alfa-2a (e.g., PEGASYS®) monotherapy is 180 μg (1.0 mL) once weekly for 48 weeks by subcutaneous (SC) administration in the abdomen or thigh.

A therapeutically effective dosage of consensus interferon alfa (e.g., INFERGEN®), particularly for treatment of chronic HCV infection, is 9 mcg TIW administered SC as a single injection for 24 weeks. At least 48 hours should elapse between doses of INFERGEN®.

Patients who tolerated previous interferon therapy and did not respond or relapsed following its discontinuation may be subsequently treated with 15 mcg of INFERGEN® TIW administered SC as a single injection for up to 48 weeks.

For patients who experience a severe adverse reaction on INFERGEN®, dosage should be withheld temporarily. If the adverse reaction does not become tolerable, therapy should be discontinued. Dose reduction to 7.5 mcg may be necessary following an intolerable adverse event.

If adverse reactions continue to occur at the reduced dosage, the physician may discontinue treatment or reduce dosage further. However, decreased efficacy may result from continued treatment at dosages below 7.5 mcg.

During subsequent treatment for 48 weeks with 15 mcg of INFERGEN®, up to 36% of patients required dose reductions in 3 mcg increments.

A therapeutically effective does of albumin-interferon-alpha (e.g., ALBUFERON®) is about 120 mcg or about 180 mcg or about 240 mcg or about 320 mcg or about 400 mcg or about 500 mcg per day subcutaneously.

A therapeutically effective dose of ribavirin (e.g., REBETROL®) depends on the patient's body weight. The recommended dose of REBETOL® is provided, below, in Table 1.

TABLE 1 Recommended Dosing Body weight REBETOL Capsules </=75 kg 2 × 200-mg capsules AM, 3 × 200-mg capsules PM daily p.o. >75 kg 3 × 200 mg capsules AM, 3 × 200 mg capsules PM daily p.o.

The recommended duration of treatment with ribavirin (e.g., REBETOL®) for patients previously untreated with interferon is 24 to 48 weeks. The duration of treatment should be individualized to the patient depending on baseline disease characteristics, response to therapy, and tolerability of the regimen. After 24 weeks of treatment, virologic response should be assessed. Treatment discontinuation should be considered in any patient who has not achieved an HCV RNA below the limit of detection of the assay by 24 weeks.

In patients who relapse following interferon therapy, the recommended duration of treatment with ribavirin (e.g., REBETOL®) is 24 weeks.

REBETOL® may be administered without regard to food, but should be administered in a consistent manner with respect to food intake.

A combination of interferon alfa-2b and ribavirin (e.g., REBETRON®) can also be administered in association with an E1-E2 peptide of the present invention (e.g., SEQ ID NOs: 3-139).

The recommended duration of REBETRON® treatment for patients previously untreated with interferon is 24 to 48 weeks. The duration of treatment should be individualized to the patient depending on baseline disease characteristics, response to therapy, and tolerability of the regimen. After 24 weeks of treatment, virologic response should be assessed. Treatment discontinuation should be considered in any patient who has not achieved an HCV RNA below the limit of detection of the assay by 24 weeks. In patients who relapse following interferon therapy, the recommended duration of treatment is 24 weeks.

The recommended dosage of a combination of ribavirin (e.g., REBETROL®) and interferon alfa-2b (e.g., INTRON-A®) depends on patient body weigh. The adult dosage regimen is set forth below in Table 2:

TABLE 2 Recommended Adult Dosing Body weight REBETOL Capsules INTRON A Injection </=75 kg 2 × 200-mg capsules AM, 3 million IU 3 times 3 × 200-mg capsules PM weekly s.c. daily p.o. >75 kg 3 × 200-mg capsules AM, 3 million IU 3 times 3 × 200-mg capsules PM weekly s.c. daily p.o.

The pediatric dosage regimen, for the combination, is set forth below in Table 3:

TABLE 3 Pediatric Dosing Body weight REBETOL Capsules INTRON A Injection 25-36 kg 1 × 200-mg capsule AM 3 million IU/m2 3 times 1 × 200-mg capsule PM weekly s.c. daily p.o. 37-49 kg 1 × 200-mg capsule AM 3 million IU/m2 3 times 2 × 200-mg capsules PM weekly s.c. daily p.o. 50-61 kg 2 × 200-mg capsules AM 3 million IU/m2 3 times 2 × 200-mg capsules PM weekly s.c. daily p.o. >61 kg Refer to adult dosing table Refer to adult dosing table

Dosage modification of REBETOL®/INTRON-A® treatment is indicated when adverse reactions are observed in the patient. For example, in patients with a history of stable cardiovascular disease, a permanent dose reduction is required if the patient's hemoglobin decreases by >1=2g/dL during any 4-week period. In addition, for these cardiac history patients, if the patient's hemoglobin remains <12g/dL after 4 weeks on a reduced dose, the patient should discontinue combination REBETOL®/INTRON-A® therapy.

It is recommended that a patient whose hemoglobin level falls below 10 g/dL have his/her REBETOL® dose reduced to 600 mg daily (1×200-mg capsule AM, 2×200-mg capsules PM). A patient whose hemoglobin level falls below 8.5 g/dL should be permanently discontinued from REBETOL®/INTRON A® therapy.

When administered in combination with REBETOL®, the recommended dose of PEG-Intron® is 1.5 micrograms/kg/week. The recommended dose of REBETOL® is 800 mg/day in 2 divided doses: two capsules (400 mg) with breakfast and two capsules (400 mg) with dinner. REBETOL® should not be used in patients with creatinine clearance <50 mL/min.

EXAMPLES

The following examples are intended to exemplify and further clarify what is the present invention and should not be construed to limit the present invention. The present invention should not be limited by any mechanism or theory presented herein. Any composition disclosed in any of the following examples along with any disclosed method is part of the present invention.

Example 1 Identification and Characterization of Inhibitory E1-E2 Peptides

Expression of HCV envelope protein. The envelope region of HCV was PCR amplified from FKI389/core3′ plasmid (Pietschmann et al., J. Virol. 76(8):4008-21 (2002)) with the primers GATCAAGCTTATGGGTTGCTCCTTTTCTATCTTC (SEQ ID NO: 140) and GATCAGATCTAGTGATAATCCGGAGTCGAACTCGATAGTC (SEQ ID NO: 141). The amplified product was digested with HindIII and XbaI and ligated into the pCMV-hygro (Invitrogen Corp.; Carlsbad, Calif.). The expression of the envelope proteins was confirmed with Western blot.

HCV pseudotype particle generation. Single cycle HCV pseudoparticles are composed of a replication-defective HIV genome and HCV envelope proteins. These pseudoparticles were made by transfecting 293T cells with 5 ug of each pNL43E-R-luc (Chen et al., J. Virol. 68(2):654-60 (1994)) and plasmid expressing HCV envelope proteins pCMV/E1-2 with profection transfection kit from Promega (Madison, Wis.). The medium was refreshed 12-24 hours after transfection and supernatant was collected 48 hours after transfection. The supernatant from a transfection of only pNL43E-R-luc was used each time as a control for non specific entry events.

Peptide library preparation. The peptide library was synthetically assembled on MBHA Rink amide or NovaSyn TG Sieber resin (at 5 umol per peptide) in 96-well microtiter plate by a MultiPep synthesizer (Intavis AG, Germany) with standard Fmoc chemistry. After successful completion of the synthesis, the resins were washed with dichloromethane five times and completely dried under vacuum in the MultiPep. The microtiter plate was removed from the MultiPep and placed on top of a 96-well collection plate (2 ml volume per well). 200 ul of concentrated (80-90%) TFA with proper scavengers such as water and triisopropylsilane were added to the microtiter wells containing the assembled peptide resin. This cleavage process was repeated three additional times with 10 min intervals. The collected cleavage solution was let to stand at room temperature for 2 hours of deprotection. Each peptide was then transferred to a 15-ml polypropylene tube containing 10 ml anhydrous ethyl ether. The tubes were spun down in a clinical centrifuge. The ether phase was carefully decanted. The precipitated peptides were taken up in water and lyophilized over night. LCMS analysis was conducted for each peptide to verify the molecular weight and assess purity. The peptides were finally dissolved in 100% DMSO and used in assays without further purification unless otherwise noted for selected bioactive peptides. Occasionally, the bioactive peptides were synthesized at larger scale (250 umol) by an ABI model 431A peptide synthesizer (Applied Biosystems, Foster City, Calif.) using the same chemistry as mentioned above.

Infection of hepatoma cell lines with HCV pseudotype particles and screen for inhibitory peptides. The hepatoma cells, Huh7, was seeded at 8000 cells in 90 ul medium in 96-well plate. In the case of the inhibitor screen, the inhibitor was incubated with cells for one hour at 37° C. and then 50 ul viral supernatant was added to the cells 24 hours after seeding. The viral stock was pre-tittered and diluted to yield approximately 10,000 to 20,000 RLU. The cells were incubated at 37° C. with 5% CO2 for additional 72 hours and were harvested for luciferase assay. Peptide stocks were prepared in 100% DMSO at 10 mg/ml and were first screened at 100 ug/ml and then a dose responsive curve was generated with 10 points 3-fold titration starting from 100 ug/ml.

The specificity of the peptides were counter-screened with AMLV and HIV pseudotyped viruses using the same infection protocol. The results of the entry assay, corresponding to the peptides tested, are shown below in Table 4.

Results.

TABLE 4 Screen of the peptide library. AMLVpp Peptides HCVpp % inh % inh Y-E-V-R-N-V-S-G-V-Y-H-V-T-N-D-C 35 18 (SEQ ID NO: 3) G-V-Y-H-V-T-N-D-C-S-N-A-S-I-V-Y −23 −6 (SEQ ID NO: 4) D-C-S-N-A-S-I-V-Y-E-A-A-D-M-I-M −4 −2 (SEQ ID NO: 5) V-Y-E-A-A-D-M-I-M-H-T-P-G-C-V-P −13 10 (SEQ ID NO: 6) I-M-H-T-P-G-C-V-P-C-V-R-E-N-N-S 3 −4 (SEQ ID NO: 7) V-P-C-V-R-E-N-N-S-S-R-C-W-V-A-L −22 14 (SEQ ID NO: 8) N-S-S-R-C-W-V-A-L-T-P-T-L-A-A-R 5 28 (SEQ ID NO: 9) A-L-T-P-T-L-A-A-R-N-A-S-V-P-T-T 53 1 (SEQ ID NO: 10) A-R-N-A-S-V-P-T-T-T-I-R-R-H-V-D −26 24 (SEQ ID NO: 11) T-T-T-I-R-R-H-V-D-L-L-V-G-A-A-A 31 −5 (SEQ ID NO: 12) V-D-L-L-V-G-A-A-A-L-C-S-A-M-Y-V 39 −13 (SEQ ID NO: 13) A-A-L-C-S-A-M-Y-V-G-D-L-C-G-S-V 39 −42 (SEQ ID NO: 14) Y-V-G-D-L-C-G-S-V-F-L-V-A-Q-L-F 16 −31 (SEQ ID NO: 15) S-V-F-L-V-A-Q-L-F-T-F-S-P-R-R-H 3 −148 (SEQ ID NO: 16) L-F-T-F-S-P-R-R-H-E-T-V-Q-D-C-N 9 16 (SEQ ID NO: 17) R-H-E-T-V-Q-D-C-N-C-S-I-Y-P-G-H 5 17 (SEQ ID NO: 18) C-N-C-S-I-Y-P-G-H-V-T-G-H-R-M-A 49 26 (SEQ ID NO: 19) G-H-V-T-G-H-R-M-A-W-D-M-M-M-N-W 43 6 (SEQ ID NO: 20) M-A-W-D-M-M-M-N-W-S-P-T-A-A-L-V 32 3 (SEQ ID NO: 21) N-W-S-P-T-A-A-L-V-V-S-Q-L-L-R-I 54 −11 (SEQ ID NO: 22) L-V-V-S-Q-L-L-R-I-P-Q-A-V-V-D-M 3 −1 (SEQ ID NO: 23) R-I-P-Q-A-V-V-D-M-V-A-G-A-H-W-G 25 −2 (SEQ ID NO: 24) D-M-V-A-G-A-H-W-G-V-L-A-G-L-A-Y 27 14 (SEQ ID NO: 25) W-G-V-L-A-G-L-A-Y-Y-S-M-V-G-N-W 50 27 (SEQ ID NO: 26) A-Y-Y-S-W-V-G-N-W-A-K-V-L-I-V-M 39 −2 (SEQ ID NO: 27) N-W-A-K-V-L-I-V-M-L-L-F-A-G-V-D 13 −1 (SEQ ID NO: 28) V-M-L-L-F-A-G-V-D-G-G-T-Y-V-T-G −22 11 (SEQ ID NO: 29) V-D-G-G-T-Y-V-T-G-G-T-M-A-K-N-T 2 −9 (SEQ ID NO: 30) T-G-G-T-N-A-K-N-T-L-G-I-T-S-L-F 41 −2 (SEQ ID NO: 31) N-T-L-G-I-T-S-L-F-S-P-G-S-S-Q-K 11 2 (SEQ ID NO: 32) L-F-S-P-G-S-S-Q-K-I-Q-L-V-N-T-N 61 −49 (SEQ ID NO: 33) Q-K-I-Q-L-V-N-T-N-G-S-W-H-I-N-R 6 13 (SEQ ID NO: 34) T-N-G-S-W-H-I-N-R-T-A-L-N-C-N-D 29 30 (SEQ ID NO: 35) N-R-T-A-L-N-C-N-D-S-L-N-T-G-F-L 23 18 (SEQ ID NO: 36) N-D-S-L-N-T-G-F-L-A-A-L-F-Y-V-H 14 15 (SEQ ID NO: 37) F-L-A-A-L-F-Y-V-H-K-F-N-S-S-G-C 23 −44 (SEQ ID NO: 38) V-H-K-F-N-S-S-G-C-P-E-R-M-A-S-C −11 −15 (SEQ ID NO: 39) G-C-P-E-R-M-A-S-C-S-P-I-D-A-F-A 35 −4 (SEQ ID NO: 40) S-C-S-P-I-D-A-F-A-Q-G-W-G-P-I-T −11 25 (SEQ ID NO: 4l) F-A-Q-G-W-G-P-I-T-Y-N-E-S-H-S-S 49 15 (SEQ ID NO: 42) I-T-Y-N-E-S-H-S-S-D-Q-R-P-Y-C-W −13 17 (SEQ ID NO: 43) S-S-D-Q-R-P-Y-C-W-H-Y-A-P-R-P-C 14 15 (SEQ ID NO: 44) C-W-H-Y-A-P-R-P-C-G-I-V-P-A-A-Q 63 −8 (SEQ ID NO: 45) P-C-G-I-V-P-A-A-Q-V-C-G-P-V-Y-C 29 −2 (SEQ ID NO: 46) A-Q-V-C-G-P-V-Y-C-F-T-P-S-P-V-V 40 −8 (SEQ ID NO: 47) Y-C-F-T-P-S-P-V-V-V-G-T-T-D-R-F 6 21 (SEQ ID NO: 48) V-V-V-G-T-T-D-R-F-G-V-P-T-Y-S-W 37 −22 (SEQ ID NO: 49) R-F-G-V-P-T-Y-S-W-G-E-N-E-T-D-V 34 33 (SEQ ID NO: 50) S-W-G-E-N-E-T-D-V-L-L-L-N-N-T-R 23 34 (SEQ ID NO: 51) D-V-L-L-L-N-N-T-R-P-P-Q-G-N-W-F 10 16 (SEQ ID NO: 52) T-R-P-P-Q-G-N-W-F-G-C-T-W-M-N-S 59 −42 (SEQ ID NO: 53) W-F-G-C-T-W-M-N-S-T-G-F-T-K-T-C 39 −15 (SEQ ID NO: 54) N-S-T-G-F-T-K-T-C-G-C-P-P-C-N-I 23 3 (SEQ ID NO: 55) T-C-G-G-P-P-C-N-I-G-G-I-C-N-K-T 53 22 (SEQ ID NO: 56) N-I-G-G-I-G-N-K-T-L-T-C-P-T-D-C 39 20 (SEQ ID NO: 57) K-T-L-T-C-P-T-D-C-F-R-K-H-P-E-A 50 13 (SEQ ID NO: 58) D-C-F-R-K-H-P-E-A-T-Y-T-K-C-G-S 26 −14 (SEQ ID NO: 59) E-A-T-Y-T-K-C-G-S-G-P-W-L-T-P-R 13 17 (SEQ ID NO: 60) G-S-G-P-W-L-T-P-R-C-L-V-H-Y-P-Y −15 −8 (SEQ ID NO: 61) P-R-C-L-V-H-Y-P-Y-R-L-W-H-Y-P-C 10 −42 (SEQ ID NO: 62) P-Y-R-L-W-H-Y-P-C-T-V-N-F-T-I-F 44 3 (SEQ ID NO: 63) P-C-T-V-N-F-T-I-F-K-V-R-M-Y-V-G −2 −30 (SEQ ID NO: 64) I-F-K-V-R-M-Y-V-G-G-V-E-H-R-L-E 19 −62 (SEQ ID NO: 65) V-G-G-V-E-H-R-L-E-A-A-C-N-W-T-R 22 34 (SEQ ID NO: 66) L-E-A-A-C-N-W-T-R-G-E-R-C-N-L-E 26 37 (SEQ ID NO: 67) T-R-G-E-R-C-N-L-E-D-R-D-R-S-E-L 13 −7 (SEQ ID NO: 68) L-E-D-R-D-R-S-E-L-S-P-L-L-L-S-T −15 1 (SEQ ID NO: 69) E-L-S-P-L-L-L-S-T-T-E-W-Q-V-L-P 10 44 (SEQ ID NO: 70) S-T-T-E-W-Q-V-L-P-C-S-F-T-T-L-P 44 18 (SEQ ID NO: 71) L-P-C-S-F-T-T-L-P-A-L-S-T-G-L-I −2 20 (SEQ ID NO: 72) L-P-A-L-S-T-G-L-I-H-L-H-Q-N-V-V 19 0 (SEQ ID NO: 73) L-I-H-L-H-Q-N-V-V-D-V-Q-Y-L-Y-G 22 5 (SEQ ID NO: 74) V-V-D-V-Q-Y-L-Y-G-I-G-S-A-V-V-S 39 44 (SEQ ID NO: 75) Y-G-I-G-S-A-V-V-S-F-A-I-K-W-E-Y 7 24 (SEQ ID NO: 76) V-S-F-A-I-K-W-E-Y-V-L-L-L-F-L-L 96 3 (SEQ ID NO: 77) E-Y-V-L-L-L-F-L-L-L-A-D-A-R-V-C 38 34 (SEQ ID NO: 78) L-L-L-A-D-A-R-V-C-A-C-L-W-M-M-L 77 16 (SEQ ID NO: 79) V-C-A-C-L-W-M-M-L-L-I-A-Q-A-E-A 16 7 (SEQ ID NO: 80) 1Percentage of cell entry inhibition of hepatitis C virus observed in the presence of the indicated peptide as compared to the uninhibited control. 2Percentage of cell entry inhibition of amphotropic murine leukemia virus observed in the presence of the indicated peptide as compared to the uninhibited control.

The data points for the dose response curve of peptide VSFAIKWEYVLLLFLL (SEQ ID NO: 77) are shown below in tables 5A (HCVpp) and 5B (HIVpp). The peptide of SEQ ID NO: 77 was first serial diluted by the factor of 3 in 100% DMSO from 100 ug/ml and then transferred to the assay plates at 100-fold dilution. The peptide was first incubated with cells and HCVpp (A) or HIVpp (B), respectively, for 30 minutes at 370 C in this experiment. Then, the medium containing the peptide was removed from the cells and virus with peptide was added to the cells. Cells were incubated for 72 hours and then were harvested for luciferase assay. Two separate trials were performed.

TABLE 5A Dose response curve data points for SEQ ID NO: 77 (HCV). Peptide conc. RLU units RLU units (log ug/ml) (1st trial) (2nd trial) 2 11 10 1.522879 22 67 1.045758 306 75 0.568636 544 500 0.091515 1044 833 −0.38561 1279 1283 −0.86273 1866 1762 −1.33985 2564 2703 −1.33985 2998 3512 −1.33985 2090 3359 1494

TABLE 5B Dose response curve data points for SEQ ID NO: 77 (HIV). Peptide conc. RLU units RLU units (log ug/ml) (1st trial) (2nd trial) 2 435408 543239 1.522879 787025 683636 1.045758 740592 744482 0.568636 561454 612198 0.091515 674698 651004 −0.38561 732652 734586 −0.86273 676303 639921 −1.33985 815312 754404

In addition, the peptide of SEQ ID NO: 77 was mutated at several positions and assayed in the HCV entry assay discussed above. The peptides generated are shown, below, in table 6 along with the results of the HCV entry assay using each peptide. Generally, the peptide of SEQ ID NO: 77 had its N-terminal three amino acids removed (13-mer) and various residues in the 13-mer were replace with alanine (Ala-scan), lysine (Lys-scan), proline (Pro-scan) or glycine (Gly-scan). The data shown in table 6 indicate the percentage of hepatitis C virus cellular entry, at 30 μM peptide concentration, as compared to the uninhibited control. The assays were performed essentially as described above.

TABLE 6 Screen of mutant peptides. % of control Sequence Description at ~30 uM A-I-K-W-E-Y-V-L-L-L-F-L-L 13-mer 17 (SEQ ID NO: 81) G-I-K-W-E-Y-V-L-L-L-F-L-L Gly-scan 22 (SEQ ID NO: 82) A-G-K-W-E-Y-V-L-L-L-F-L-L Gly-scan 9 (SEQ ID NO: 83) A-I-K-W-G-Y-V-L-L-L-F-L-L Gly-scan 19 (SEQ ID NO: 84) A-I-K-W-E-Y-V-L-L-L-F-L-L 13-mer 20 (SEQ ID NO: 85) A-I-K-W-E-Y-V-L-L-L-F-L-L 13-mer 13 (SEQ ID NO: 86) A-A-K-W-E-Y-V-L-L-L-F-L-L Ala-scan 14 (SEQ ID NO: 87) A-I-A-W-E-Y-V-L-L-L-F-L-L Ala-scan 9 (SEQ ID NO: 88) A-I-K-A-E-Y-V-L-L-L-F-L-L Ala-scan 8 (SEQ ID NO: 89) A-I-K-W-A-Y-V-L-L-L-F-L-L Ala-scan 13 (SEQ ID NO: 90) A-I-K-W-E-A-V-L-L-L-F-L-L Ala-scan 16 (SEQ ID NO: 91) A-I-K-W-E-Y-A-L-L-L-F-L-L Ala-scan 17 (SEQ ID NO: 92) A-I-K-W-E-Y-V-A-L-L-F-L-L Ala-scan 13 (SEQ ID NO: 93) A-I-K-W-E-Y-V-L-A-L-F-L-L Ala-scan 17 (SEQ ID NO: 94) A-I-K-W-E-Y-V-L-L-L-A-L-L Ala-scan 8 (SEQ ID NO: 95) A-I-K-W-E-Y-V-L-L-L-F-A-L Ala-scan 24 (SEQ ID NO: 96) A-I-K-W-E-Y-V-L-L-L-F-L-A Ala-scan 24 (SEQ ID NO: 97) A-I-K-W-E-Y-V-L-L-F-L-L-L 13-mer 10 (SEQ ID NO: 98) P-I-K-W-E-Y-V-L-L-L-F-L-L Pro-scan 21 (SEQ ID NO: 99) A-I-K-W-E-Y-V-L-L-L-F-L-L 13-mer 6 (SEQ ID NO: 100) K-I-K-W-E-Y-V-L-L-L-F-L-L Lys-scan 4 (SEQ ID NO: 101) A-K-K-W-E-Y-V-L-L-L-E-L-L Lys-scan 5 (SEQ ID NO: 102) A-I-K-W-K-Y-V-L-L-L-F-L-L Lys-scan 1 (SEQ ID NO: 103)

Example 2 The Combinational Effects of an Anti-CD81 Antibody and the Peptide of SEQ ID NO: 77 on HCV Pseudoparticle Entry

This example demonstrated that the peptide of SEQ ID NO: 77 can enhance the potency of an anti-CD81 antibody and reduce the amount of anti-CD81 antibody that is needed to achieve HCV entry inhibition. The anti-CD81 antibody and the peptide were titrated in duplicates in a 96-well formatted matrix and tested in an HCV pseudoparticle entry assay. The assays were carried out essentially as described above. The data are shown below in the table 7.

TABLE 7 Results of HCV pseudoparticle entry assay in the presence of the peptide of SEQ ID NO: 77 and an anti-CD81 antibody. CD81 peptide antibody ug/ml 100 100 25 25 6.25 6.25 1.563 1.563 ug/ml RLU RLU RLU RLU RLU RLU RLU RLU RLU 10 RLU 11 54 9 11 59 8 12 8 1 RLU 15 1322 60 97 62 183 82 69 0.5 RLU 425 2378 465 547 639 379 886 1421 0.25 RLU 895 2056 1329 449 1716 1286 1177 1863 0.125 RLU 2022 2646 1972 1953 3028 3033 5729 3990 0.063 RLU 2490 1686 2866 3310 4473 4063 6595 4999 0.031 RLU 3956 2902 2898 4125 5178 5950 7372 6792 0.016 RLU 3651 3704 6733 3753 5114 3947 10490 8055 0.008 RLU 3467 3174 7394 5876 8317 7338 7426 7750 0.004 RLU 5596 2937 6870 4968 7406 7571 13281 7992 0.002 RLU 3192 3875 6206 5485 7305 7703 10717 9978 0.002 RLU 2317 4082 5297 5697 5277 8751 11221 9543 CD81 antibody 0.391 0.391 0.098 0.098 0 0 0 0 ug/ml RLU RLU RLU RLU RLU RLU RLU RLU 10 11 9 16 7 106 9 85 45 1 392 312 298 141 779 303 1005 470 0.5 505 1120 1402 491 1608 1338 1686 1356 0.25 3172 1404 2168 2740 6099 4330 9891 5351 0.125 5847 4578 3503 7501 14512 9507 15468 8386 0.063 10137 7624 10587 9989 17164 12430 16250 12146 0.031 9934 8742 15376 12424 16998 13731 19768 16741 0.016 12692 9424 15036 12663 17381 17712 17123 15484 0.008 15805 9670 17882 13398 19216 16711 23553 15269 0.004 14122 13155 18445 6152 18053 19403 18423 16197 0.002 13906 13219 20304 13319 10409 14130 17576 15204 0.002 9828 12349 14565 17382 15086 16310 13542 17922 RLU = luciferase units (relative light units).

Dose response curves of anti-CD81 antibody with various amounts of peptide were plotted vs. RLU, and the results are summarized below. The data indicate that the concentrations for anti-CD81 antibody to achieve 50% and 90% inhibition were decreased in the presence of the increasing amount of the peptide. When the peptide was present at 10-30% of its IC50 concentration, the IC50 for anti-CD81 was reduced by 3 to 6-fold. Similarly, when the peptide was present at 1% to 25% of its IC75 concentration, the IC75 for the anti-CD81 antibody was reduced by 3 to 9 fold. Thus, the peptide enhances the potency of the anti-CD81 antibody.

TABLE 8 IC50 and IC90 of the anti-CD81 antibody in the presence of increasing concentrations of peptide of SEQ ID NO: 77. CD81 + pp75 ug/ml IC50 IC90 0.000 0.214 0.639 0.000 0.189 0.482 0.098 0.090 0.319 0.391 0.063 0.378 1.563 0.024 0.371 6.250 0.253 25.000 0.139 100.000 0.266

Example 3 Amino Acid Sequence Requirement for Peptide Activity

To determine which amino acids are required to inhibit HCVpp entry, sequential deletions were generated at the amino and carboxy terminus of the peptide of SEQ ID NO: 77 (Table 9). Sequential deletion of the first six amino acids from the amino terminus retained 80 to 90% of inhibition activity; however, a series removal of the six amino acids from the carboxy terminus lost approximately 50% activity. The sensitivity to the carboxy terminus deletions indicates the importance of the residues from the transmembrane domain. The peptide, A-I-K-W-E-Y-V-L-L-L-F-L-L (SEQ ID NO: 106 or 81), retained about 90% activity and is three amino acids shorter than the peptide of SEQ ID NO: 77.

TABLE 9 Truncated peptides.¢ Peptides Comments HCVpp % inh V-S-F-A-I-K-W-E-Y-V-L-L-L-F-L-L wild type (SEQ ID NO: 77)   S-F-A-I-K-W-E-Y-V-L-L-L-F-L-L N-terminal truncation 91 (SEQ ID NO: 104)     F-A-I-K-W-E-Y-V-L-L-L-F-L-L N-terminal truncation 95 (SEQ ID NO: 105)       A-I-K-W-E-Y-V-L-L-L-F-L-L N-terminal truncation 92 (SEQ ID NO: 106)         I-K-W-E-Y-V-L-L-L-F-L-L N-terminal truncation 88 (SEQ ID NO: 107)           K-W-E-Y-V-L-L-L-F-L-L N-terminal truncation 78 (SEQ ID NO: 108)             W-E-Y-V-L-L-L-F-L-L N-terminal truncation 82 (SEQ ID NO: 109) V-S-F-A-I-K-W-E-Y-V-L-L-L-F-L-L wild type (SEQ ID NO: 110) V-S-F-A-I-K-W-E-Y-V-L-L-L-F-L C-terminal truncation 63 (SEQ ID NO: 111) V-S-F-A-I-K-W-E-Y-V-L-L-L-F C-terminal truncation 49 (SEQ ID NO: 112) V-S-F-A-I-K-W-E-Y-V-L-L-L C-terminal truncation 49 (SEQ ID NO: 113) V-S-F-A-I-K-W-E-Y-V-L-L C-terminal truncation 44 (SEQ ID NO: 114) V-S-F-A-I-K-W-E-Y-V-L C-terminal truncation 17 (SEQ ID NO: 115) V-S-F-A-I-K-W-E-Y-V C-terminal truncation 60 (SEQ ID NO: 116) ¢Peptides were dissolved at 5 mM in 100% DMSO and tested at 50 uM in 1% DMSO.

Compared to the wild type 13-mer peptide (SEQ ID NO: 106 or 81), the retro peptide of SEQ ID NO: 106 (i.e., LLFLLLVYEWKIA (SEQ ID NO: 107)) and retro-inverso peptide of SEQ ID NO: 106 (i.e., LLFLLLVYEWKIA (SEQ ID NO: 107) with all D-amino acids) peptides completely lost activity, indicating that the overall hydrophobic nature of the retro and retro-inverso peptides is not enough for activity and the sequential order of certain key amino acids plays a leading role. The inverso peptides (SEQ ID NO: 106 with all D-amino acids), however, retained partial but weakened activity.

TABLE 10 Dose response curve for HCV entry at various concentrations of inverso, retro and retro-inverso peptides¥ nM of peptide inverso inverso inverso retro retro 30000 30 68 12 54 19 34 90 96 80 73 10000 40 44 41 46 27 52 54 99 55 79 300 78 62 70 77 46 41 66 79 119 69 100 67 74 106 84 60 82 101 79 99 72 30 120 94 91 86 72 45 58 93 92 94 10 115 97 99 74 86 89 95 93 81 52 3 109 86 108 74 80 90 99 121 76 62 1 98 87 73 123 99 88 86 97 81 94 0.3 118 102 110 112 73 69 98 86 95 88 0.1 139 111 90 93 117 94 87 114 83 82 0.1 116 69 91 88 68 101 93 109 96 90 0.1 122 92 109 112 124 108 98 100 107 107 nM of retro- retro- retro- SEQ ID peptide retro inverso inverso inverso NO: 81 30000 104 110 107 87 103 125 107 95 30 26 10000 34 63 32 55 80 70 90 73 22 34 300 54 62 57 71 64 79 94 115 39 31 100 65 62 99 62 91 102 142 81 69 24 30 45 64 68 62 76 113 121 110 51 38 10 68 77 66 100 76 84 128 104 70 71 3 77 66 88 68 78 92 86 98 82 66 1 51 88 83 72 91 115 106 115 94 114 0.3 88 91 91 81 74 99 126 117 90 105 0.1 72 67 81 100 82 113 98 97 103 79 0.1 90 102 85 99 92 95 103 88 86 77 0.1 92 116 106 110 106 107 112 97 104 134 ¥Numbers represent viral entry in the presence of the indicated peptide as a percentage of the uninhibited control.

TABLE 11 Dose response curve for HCV entry at various peptide concentrations.£ uM of scrambled SEQ scrambled SEQ scrambled SEQ peptide ID NO: 138 ID NO: 138 ID NO: 138 SEQ ID NO: 81 SEQ ID NO: 85 30 7047 5340 7406 5158 5107 5826 990 667 907 406 10 9469 9957 12877 7340 8434 6401 2584 3853 2865 3393 3.333333 7729 11535 12632 8653 8028 10288 1152 2683 2490 2239 1.111111 6456 7681 9683 9482 9053 7359 2065 2449 2918 4380 0.3703704 7620 8597 9180 5716 8494 8291 2133 2617 3430 3658 0.1234568 9257 8961 11382 10746 7229 6598 1665 1759 2304 5077 0.04115226 12287 13159 13838 13589 10645 10230 3138 5487 6444 7203 0.01371742 13068 15343 14994 12097 11663 11748 4392 3561 7335 8987 0.01371742 15761 17272 24285 16582 13218 17842 13679 16041 14360 13526 0.01371742 20329 16998 20395 15327 17640 13895 12493 16554 13698 15168 uM of peptide SEQ ID NO: 86 SEQ ID NO: 98 SEQ ID NO: 100 SEQ ID NO: 103 30 1899 1645 1938 2251 1262 1073 2946 4314 10 2867 1216 2186 2352 1538 1360 2732 4879 3.333333 2746 2356 2647 2056 4131 3158 3098 4136 1.111111 3652 3574 3285 2555 3041 2870 3136 4198 0.3703704 5748 3127 2830 5272 4161 2767 2800 5362 0.1234568 6445 3869 4583 6873 4241 3590 5848 7916 0.04115226 8349 8612 6486 6677 7066 7585 10722 14319 0.01371742 10981 12701 9747 12623 9526 11368 13380 14899 0.01371742 17159 18427 16027 17299 13637 13231 14290 13421 0.01371742 19121 16287 19079 17575 14358 15831 19490 13601

TABLE 12 Dose response curve for AMLV entry at various peptide concentrations.£ uM of scrambled SEQ scrambled SEQ scrambled SEQ peptide ID NO: 106 ID NO: 106 ID NO: 106 SEQ ID NO: 81 SEQ ID NO: 85 30 87785 60410 83010 77083 85612 79287 86600 64937 83257 72040 10 70668 50409 97446 87834 101416 64206 153900 69448 172763 129890 3.333333 50674 41738 60859 50020 51236 49388 62658 51388 118949 92146 1.111111 49084 39443 53349 51341 46069 46019 54439 53032 48628 42996 0.3703704 48158 45212 49139 49219 51358 50551 48861 55221 43073 44659 0.1234568 39298 28711 46132 49974 42958 51726 51789 49401 44739 41483 0.04115226 43854 39721 46556 44480 40734 47413 40104 51496 42604 32390 0.01371742 49759 43879 46668 38054 46277 49423 46029 39655 43767 55532 0.01371742 37110 39504 40808 46160 44415 52102 41740 42548 44812 32138 0.01371742 37232 42195 45601 44163 52921 49326 45150 45203 40068 41172 uM of peptide SEQ ID NO: 86 SEQ ID NO: 98 SEQ ID NO: 100 SEQ ID NO: 103 30 76174 71333 81103 70832 76883 56072 116018 90397 10 70573 72462 111324 59835 135633 74847 182409 156275 3.3333 53350 39047 59984 48300 70934 59407 60851 69213 1.1111 40296 43593 45508 44147 45723 43633 49626 41878 0.3704 43543 47987 42748 48710 42008 39907 49219 41944 0.1235 37163 47412 45455 45030 46954 44234 53044 67129 0.0412 42084 47952 41463 41610 39784 45324 45898 42568 0.0137 42295 44556 44492 44531 45888 43209 44656 41299 0.0137 43059 48854 47406 49869 52210 102903 45140 43684 0.0137 37783 49462 51067 61382 41952 46766 39179 81198 £Numbers represent luciferase units observed in the viral entry assays conducted. SEQ ID Nos: 81, 85, 86, 98 and 100 represent the identical wild type 13-mer sequence from independent synthesis.

Scrambled 13-mer peptides were clearly different from the wild type 13-mer but yet retained some HCVpp-specific inhibition. The fact that the scrambled 13-mer LKLFEVYLILWLA (SEQ ID NO: 138) still showed some activity, though weakened, implies that what matters for the activity is either the overall hydrophobicity of the peptide and/or the preeminent importance of perhaps as few as two or three of the residues, or the accumulative small contributions from residues in the right places. However, as noted above, since the equally hydrophobic retro and retro-inverso peptides were inactive, and as we shall see below, the contributions to activity are most likely from many residues, thus it is more likely that a significant portion of anti-viral activity comes from residues that happen to be at the approximate right places in the overall scrambled context. The amino acid scanning experiments, below, reaffirmed the notion of collective contributions by many residues.

Gly scanning: Glycine substitutions not only eliminated the side chains, but they were expected to introduce much greater local backbone conformational flexibilities. This enhanced conformational freedom can effectively dilute the presence of bioactive conformations, leading to weaker activities. Peptides of SEQ ID NOs: 82-84 and 111 are similarly active or slightly better than wild-type, others were less active. It appears that the N-terminal AIKWEY (SEQ ID NO: 139) sequence can tolerate four (A/I/E/Y) of six substitutions, whereas the Leu-rich carboxy terminal segment is less forgiving to glycine replacement.

The data generated in the glycine scanning experiments is set forth below in Table 13.

TABLE 13 Glycine scanning experiments SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID uM of NO: 81 NO: 82 NO: 83 NO: 109 NO: 110 peptide (% control) (% control) (% control) (% control) (% control) 30 30 26 18 29 18 15 50 28 3 23 10 22 34 33 29 27 89 66 80 24 18 0.3 39 31 33 25 17 56 40 69 51 54 0.1 69 24 33 27 19 47 60 77 61 82 0.03 51 38 46 41 43 62 61 72 87 71 0.01 70 71 53 43 76 56 84 111 77 81 0.003 82 66 71 80 51 66 99 139 102 65 0.001 94 114 90 66 70 67 93 96 109 91 0.0003 90 105 68 71 94 83 94 100 107 97 0.0001 103 79 62 61 75 94 86 103 84 100 0.00003 86 77 74 88 98 80 88 110 119 104 0.00003 104 134 118 120 128 94 93 109 87 90 SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID uM of NO: 84 NO: 111 NO: 112 NO: 113 NO: 114 peptide (% control) (% control) (% control) (% control) (% control) 30 22 38 10 22 24 37 41 147 118 19 10 10 40 22 21 34 51 38 36 30 28 0.3 37 57 36 42 51 61 46 56 51 34 0.1 41 39 50 46 67 62 78 72 46 64 0.03 62 52 42 52 64 79 76 78 93 101 0.01 72 53 49 81 71 69 101 95 82 113 0.003 70 70 56 79 83 121 94 116 92 102 0.001 74 73 65 92 100 88 101 94 101 84 0.0003 91 81 111 59 71 88 87 121 84 92 0.0001 62 75 81 86 103 112 89 92 93 85 0.00003 118 84 82 99 112 74 103 115 85 89 0.00003 87 110 114 106 111 103 80 101 111 115

Ala scanning: Contrary to glycine, devoid of any side chain, alanine still maintains similar backbone conformational rigidity as other L-amino acids. Most alanine mutants are similar to the wild-type, except for the peptide of SEQ ID NO: 115. These data suggested that the expression of viral entry blocking activity is contributed largely collectively by many constituent residues, except somewhat more significantly by the leucine amino terminal to phenylalanine.

The data generated in the alanine scanning experiments is set forth below in Table 14.

TABLE 14 Alanine scanning experiments SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID nM of NO: 85 NO: 86 NO: 87 NO: 88 NO: 89 NO: 90 NO: 91 peptide (% control) (% control) (% control) (% control) (% control) (% control) (% control) 30000 70 103 15 27 11 6 8 45 4 12 29 4 12 18 10000 13 18 27 25 15 23 27 32 24 17 40 14 30 15 300 18 22 25 30 16 13 42 45 24 27 55 55 17 27 100 45 50 48 33 19 15 37 57 41 38 51 43 35 32 30 62 57 59 60 42 51 53 66 66 71 69 63 49 48 10 90 107 69 67 45 57 56 60 63 57 66 46 59 66 3 71 88 90 118 92 68 81 93 63 76 106 81 86 110 1 95 83 80 102 110 93 100 69 85 96 66 98 120 102 0.3 79 96 96 133 84 75 91 101 71 64 73 94 81 84 0.1 79 81 86 90 90 108 96 85 84 76 105 98 101 73 0.1 101 74 87 103 113 90 87 108 104 95 82 106 107 89 0.1 113 112 103 108 93 104 96 109 96 105 116 96 93 111 SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID nM of NO: 92 NO: 93 NO: 94 NO: 115 NO: 95 NO: 96 NO: 97 peptide (% control) (% control) (% control) (% control) (% control) (% control) (% control) 30000 5 15 32 32 14 28 36 0 7 23 31 31 53 50 10000 12 27 41 32 36 34 11 38 25 15 33 20 19 37 300 21 30 32 27 42 33 64 40 18 13 36 26 30 47 100 32 25 47 42 45 41 54 53 35 53 24 41 46 51 30 42 64 58 51 58 48 88 72 75 35 63 67 55 80 10 82 59 71 67 79 86 105 78 64 47 58 73 79 69 3 68 99 78 87 58 89 102 86 85 52 76 85 72 92 1 90 80 85 87 81 82 66 93 91 72 124 78 76 92 0.3 85 61 86 105 104 92 82 91 62 70 91 98 72 102 0.1 73 115 91 83 99 96 83 118 89 82 95 86 80 101 0.1 82 89 80 104 79 101 108 124 114 76 100 96 105 102 0.1 99 130 134 81 115 105 81 86 113 97 72 132 95 97

Pro scanning: Unlike the conformationally permissive glycine, proline is far more restrictive due to the side chain to backbone nitrogen ring closure. Introducing praline into the peptide was aimed at creating local contortion of the peptide backbone and probing if a less aggregating and more water soluble but active mutant peptide can emerge. Only the very amino terminal alanine to proline mutant (SEQ ID NO: 99) was similar to the wild-type peptide, others are less active. This suggested that local backbone contortion would ablate activity.

The data generated in the proline scanning experiments is set forth below in Table 15.

TABLE 15 Proline scanning experiments. SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID nM of NO: 98 NO: 99 NO: 116 NO: 117 NO: 118 NO: 119 NO: 120 peptide (% control) (% control) (% control) (% control) (% control) (% control) (% control) 30000 19 6 2 9 2 5 49 4 24 33 28 17 38 42 10000 27 24 24 16 15 19 54 31 16 41 21 7 24 24 300 21 29 21 29 18 34 48 41 38 29 40 47 33 39 100 31 41 21 34 49 28 38 54 47 53 40 46 39 39 30 41 35 28 36 29 46 58 71 63 93 39 79 72 40 10 47 50 45 53 61 58 75 75 116 83 54 48 73 76 3 86 52 68 56 68 86 89 66 108 84 94 62 66 76 1 93 94 75 89 81 93 68 87 74 99 72 68 91 75 0.3 90 68 84 80 86 76 66 117 101 92 78 77 90 75 0.1 85 77 64 84 89 74 128 93 80 87 93 76 82 78 0.1 86 109 108 70 98 105 91 97 103 108 87 85 102 80 0.1 109 95 101 121 120 77 144 68 110 79 102 126 101 117 SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID nM of NO: 121 NO: 122 NO: 123 NO: 124 NO: 125 NO: 126 NO: 127 peptide (% control) (% control) (% control) (% control) (% control) (% control) (% control) 30000 32 41 51 23 31 49 98 67 68 59 55 80 57 29 10000 50 23 54 43 61 51 85 60 73 54 43 44 29 27 300 42 30 76 53 41 44 85 68 63 76 72 85 63 72 100 44 63 75 59 72 61 66 83 60 58 68 81 77 74 30 70 82 70 113 74 75 48 71 80 102 83 82 78 83 10 75 66 85 67 78 76 67 102 97 109 86 76 102 74 3 85 87 106 80 92 78 80 86 85 82 98 89 66 53 1 76 110 73 100 108 77 89 102 75 99 88 93 71 85 0.3 68 84 105 83 80 91 74 88 104 104 88 91 84 81 0.1 77 95 81 70 78 81 87 98 91 99 83 66 90 86 0.1 91 115 89 86 104 86 89 104 111 92 89 87 90 75 0.1 104 90 115 111 117 94 109 97 95 101 104 119 122 113

Lys scanning: Lysine was positively charged under the assay condition. Lysine scanning was aimed at reversing the local hydrophobic character and assessing the consequence to potency. Similar to glycine scanning, the amino terminal substitutions were more tolerant; thus peptides of SEQ ID NOs: 101, 102 and 129 were similar or slightly more potent than the wild-type peptide, while others were less potent than the wild-type peptide.

The data generated in the lysine scanning experiments is set forth below in Table 16.

TABLE 16 Lysine scanning experiments SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID nM of NO: 100 NO: 101 NO: 102 NO: 103 NO: 128 NO: 103 NO: 130 peptide (% control) (% control) (% control) (% control) (% control) (% control) (% control) 30000 6 22 7 1 7 2 2 18 1 9 9 2 26 4 10000 5 20 2 18 17 7 9 23 4 7 6 17 38 3 300 32 29 26 29 29 23 30 29 28 22 15 19 33 19 100 31 19 26 25 16 25 28 27 46 52 15 27 58 24 30 53 44 37 28 33 41 29 29 55 59 27 35 74 75 10 62 58 54 57 67 62 84 67 92 63 43 50 90 102 3 61 83 52 77 70 66 80 81 81 84 91 81 95 81 1 76 68 65 90 96 83 104 61 72 99 97 85 114 89 0.3 88 75 96 103 141 107 93 79 88 64 104 83 103 100 0.1 110 85 83 107 104 102 117 92 86 80 80 93 122 84 0.1 115 83 77 104 89 108 104 77 102 107 92 87 106 96 0.1 92 111 119 101 98 106 131 88 103 87 101 120 105 93 SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID SEQ ID nM of NO: 131 NO: 132 NO: 133 NO: 134 NO: 135 NO: 136 NO: 137 peptide (% control) (% control) (% control) (% control) (% control) (% control) (% control) 30000 30 44 42 41 16 42 56 45 29 37 26 62 27 9 10000 16 14 49 50 24 39 60 47 43 57 34 68 23 28 300 29 42 46 44 41 56 75 68 70 87 81 84 54 77 100 49 62 59 62 75 60 89 87 107 75 98 84 68 74 30 71 68 71 89 80 68 101 76 89 90 65 84 78 94 10 74 66 54 56 77 113 105 77 104 79 80 71 100 89 3 71 85 78 75 95 79 94 92 74 67 76 98 82 97 1 85 68 67 79 96 82 128 81 84 76 71 90 80 106 0.3 105 81 100 71 108 78 104 85 99 72 103 70 58 92 0.1 97 80 73 89 78 85 80 96 63 96 67 81 72 123 0.1 97 94 95 92 96 101 108 75 74 85 82 94 80 82 0.1 96 113 117 96 87 116 138 79 129 112 116 108 103 135

The peptides generated in the Ala-, Lys- and Pro-scanning experiments are set forth below in table 17.

TABLE 17 Peptides generated in Ala-, Lys- and Pro- scanning experiments Amino acid sequence Sequence identifier A-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 81 G-I-K-W-E-Y-V-L-L-L-E-L-L SEQ ID NO: 82 A-G-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 83 A-I-G-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 109 A-I-K-G-E-Y-V-L-L-L-E-L-L SEQ ID NO: 110 A-I-K-W-G-Y-V-L-L-L-F-L-L SEQ ID NO: 84 A-I-K-W-E-G-V-L-L-L-F-L-L SEQ ID NO: 111 A-I-K-W-E-Y-G-L-L-L-F-L-L SEQ ID NO: 112 A-I-K-W-E-Y-V-L-L-L-F-G-L SEQ ID NO: 113 A-I-K-W-E-H-V-L-L-L-F-L-G SEQ ID NO: 114 A-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 85 A-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 86 A-A-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 87 A-I-A-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 88 A-I-K-A-E-Y-V-L-L-L-F-L-L SEQ ID NO: 89 A-I-K-W-A-Y-V-L-L-L-F-L-L SEQ ID NO: 90 A-I-K-W-E-A-V-L-L-L-F-L-L SEQ ID NO: 91 A-I-K-W-E-Y-A-L-L-L-F-L-L SEQ ID NO: 92 A-I-K-W-E-Y-V-A-L-L-F-L-L SEQ ID NO: 93 A-I-K-W-E-Y-V-L-A-L-F-L-L SEQ ID NO: 94 A-I-K-W-E-Y-V-L-L-A-E-L-L SEQ ID NO: 115 A-I-K-W-E-Y-V-L-L-L-A-L-L SEQ ID NO: 95 A-I-K-W-E-Y-V-L-L-L-F-A-L SEQ ID NO: 96 A-I-K-W-E-Y-V-L-L-L-F-L-A SEQ ID NO: 97 A-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 98 P-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 99 A-P-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 116 A-I-P-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 117 A-I-K-P-E-Y-V-L-L-L-F-L-L SEQ ID NO: 118 A-I-K-W-P-Y-V-L-L-L-F-L-L SEQ ID NO: 119 A-I-K-W-E-P-V-L-L-L-F-L-L SEQ ID NO: 120 A-I-K-W-E-Y-P-L-L-L-F-L-L SEQ ID NO: 121 A-I-K-W-E-Y-V-P-L-L-F-L-L SEQ ID NO: 122 A-I-K-W-E-Y-V-L-P-L-F-L-L SEQ ID NO: 123 A-I-K-W-E-Y-V-L-L-P-F-L-L SEQ ID NO: 124 A-I-K-W-E-Y-V-L-L-L-P-L-L SEQ ID NO: 125 A-I-K-W-E-Y-V-L-L-L-F-P-L SEQ ID NO: 126 A-I-K-W-E-Y-V-L-L-L-F-L-P SEQ ID NO: 127 A-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 100 K-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 101 A-K-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 102 A-I-K-W-E-Y-V-L-L-L-F-L-L SEQ ID NO: 103 A-I-K-K-E-Y-V-L-L-L-F-L-L SEQ ID NO: 128 A-I-K-W-K-Y-V-L-L-L-F-L-L SEQ ID NO: 129 A-I-K-W-E-K-V-L-L-L-F-L-L SEQ ID NO: 130 A-I-K-W-E-Y-K-L-L-L-F-L-L SEQ ID NO: 131 A-I-K-W-E-Y-V-K-L-L-F-L-L SEQ ID NO: 132 A-I-K-W-E-Y-V-L-K-L-F-L-L SEQ ID NO: 133 A-I-K-W-E-Y-V-L-L-K-F-L-L SEQ ID NO: 134 A-I-K-W-E-Y-V-L-L-L-K-L-L SEQ ID NO: 135 A-I-K-W-E-Y-V-L-L-L-F-K-L SEQ ID NO: 136 A-I-K-W-E-Y-V-L-L-L-F-L-K SEQ ID NO: 137

Taken together, the scanning data, establish that the amino terminal segment (AIKWE) (SEQ ID NO: 139) was more tolerant to substitutions whereas the carboxy terminal hydrophobic segment was less so. The potency was contributed collectively by many residues (as seen in alanine scanning). Peptide backbone contortion (as in proline scanning) or conformational de-constraining (as in glycine scanning) was not well tolerated. Perturbing the hydrophobic character in the carboxy terminal segment is also detrimental to activity (as seen with lysine scanning).

Example 3 Steps of HCVpp Entry Affected by the Peptide

To identify if the peptides of the invention bind to the cells and therefore inhibit entry, anti-CD81 antibody or peptide (SEQ ID NO: 77) was tested at four conditions:

    • (A) peptide or anti-CD81 antibody was pre-incubated with cells and virus separately, cells were washed and virus together with peptide was added;
    • (B) peptide or anti-CD81 antibody was pre-incubated with virus only first and then virus together with peptide was added to the cells;
    • (C) peptide or anti-CD81 antibody was preincubated with cells only and washed off, and then virus was added; and
    • (D) same as C except that virus was washed off after 1 hour of incubation.

Realizing that during virus incubation, the pre-bond peptide or antibody could dissociate from the cells and therefore lose the effects on HCVpp entry, virus was removed at time points from 15 minutes to 4 hours and we compared the inhibition effects to that from leaving viruses on until harvest for luciferase. As shown in the data of table 18, when virus was removed at earlier time points, stronger inhibition was observed compared to leaving virus on until harvest.

TABLE 18 Results of time course wherein virus was removed at various points.* anti-CD81 ug/ml 5 min 15 min 30 min 1 hr 2 hr 3 hr 4 hr leaving on no wash off 10 63 60 19 29 21 25 25 35 31 67 45 48 61 88 29 73 0 1 3.3333 54 27 34 41 36 33 27 34 32 59 54 45 58 65 61 73 4 3 1.1111 36 56 87 57 25 46 11 61 51 56 63 63 72 90 54 83 6 2 0.3704 68 98 55 59 75 71 27 43 61 86 89 71 76 112 67 95 22 22 0.1235 25 59 39 33 50 32 39 21 45 42 66 52 50 41 74 98 99 122 0.0412 22 27 69 37 89 86 52 49 53 78 58 69 62 51 72 111 88 101 0.0137 36 56 79 64 71 93 33 49 60 64 62 87 65 74 69 110 106 124 0.0046 85 27 55 66 61 72 65 66 57 73 71 87 65 53 71 112 84 97 0.0015 83 67 142 104 109 122 72 71 87 79 103 91 99 104 80 118 117 97 0.0005 127 117 68 74 75 92 88 74 63 91 87 91 92 79 77 114 94 92 0.0002 80 79 89 75 90 120 145 78 119 119 82 108 126 97 74 108 0.0002 95 145 138 98 89 101 94 83 70 92 100 110 94 83 93 124 *Numbers represent the viral entry observed at each time point and antibody concentration tested as a percentage of the uninhibited control

The majority of difference seen before the 4-hour time-point is in line with the model wherein most of the entry events occur during the first four hours (data not shown). As expected, the RLU signal was reduced at earlier time points and lead to higher variation of the assay. In general, when anti-CD81 antibody was removed, the earlier removal of virus seemed to correlate with a greater inhibition, indicating dissociation of anti-CD81 antibody from cells. In anticipating similar phenomena might exist with the peptide, we applied the one hour virus incubation in the following studies. In tables 19-22, condition D was identical to condition C except that virus was removed after 1 hour of incubation. The dose-response curves from condition A superimposed that from condition B, and little inhibition was observed at condition C, indicating the pre-bond antibody or peptide on cell offered no advantages in blocking HCVpp entry, or alternatively, the affinity of peptide and antibody to cells is low and can dissociate from cells rather quickly. The experiments performed under condition D suggested that the latter might be the case, in that the earlier the infection was terminated, there seemed to be a greater inhibition effect from the prebond peptide and antibody. Anti-CD81 antibody binds to CD81 expressed on the cells and therefore blocks HCVpp entry. Comparing to anti-CD81 antibody, the peptide exhibited more inhibition when pre-incubated with cells at 37° C. and 4° C. in the wash-off experiment, indicating that it most likely interacted with the cells and inhibited HCVpp entry.

The data in tables 19 and 20 were generated in assays conducted at 37° C. The date generated in tables 21 and 22 were generated in assays conducted at 4° C. The numbers in each table (19-22) represent the luciferase units observed in the entry assay under each indicated condition.

TABLE 19 Wash off study of anti-CD-81 antibody at 37° C. anti-CD81 antibody (ug/ml) condition A condition B condition C condition D 10 179 609 885 1378 35920 35648 5002 1964 3.333333 1923 1259 1481 1643 39146 32572 2063 3976 1.111111 2437 719 3672 1402 35842 33312 3158 2386 0.3703704 9829 9696 9224 11247 39546 36517 1192 3805 0.1234568 43590 53450 53230 41923 41983 46677 3801 8524 0.04115226 38615 44249 45687 40315 51423 45565 2736 4057 0.01371742 46405 54468 38470 48976 38356 42778 2755 5184 0.004572474 36968 42764 46670 44647 38239 42076 3899 4580 0.004572474 51599 42409 56802 46508 41564 49023 3482 7151 0.004572474 41187 40579 34004 46112 38135 44170 5511 6649

TABLE 20 Wash off study of peptide (SEQ ID NO: 77) at 37° C. peptide V-2b (SEQ ID NO: 77) (ug/ml) condition A condition B condition C condition D 100.0000 9216 7419 8074 4093 19088 14073 780 1531 33.3333 6467 6356 7146 4501 23383 19309 2351 2058 11.1111 12402 6768 10212 6902 28818 33022 3302 3259 3.7037 15025 10613 15213 12233 31857 32265 2974 3707 1.2346 21301 21733 25204 14452 36450 40467 2078 4828 0.4115 28106 25559 26979 31819 40225 43467 3257 2555 0.1372 44159 41959 43468 36136 34241 44163 2443 4834 0.0457 50460 53231 47929 53377 36956 46680 3531 2785 0.0046 74772 76125 65028 60343 40529 45271 4566 6407 0.0046 57485 58523 57349 51858 37116 32831 3984 2714

TABLE 21 Wash off study of anti-CD-81 antibody at 4° C. anti-CD81 antibody (ug/ml) condition A condition B condition C 10 42 242 78 37 6720 8027 3.3333 14 135 382 470 11036 10783 1.1111 1733 253 1815 706 10112 11783 0.3704 1885 1046 2988 1939 9220 18281 0.1235 6160 5063 7111 5662 11651 11051 0.0412 11764 8052 16527 11566 10723 11422 0.0137 12944 10622 13979 15817 7207 12271 0.0046 10693 11365 12812 11119 11552 15524 0.0046 9969 11238 15082 15072 12008 9342 0.0046 15243 12178 10078 12548 8921 9666

TABLE 22 Wash off study of peptide (SEQ ID NO: 77) at 4° C. peptide V-2b (SEQ ID No: 77) (ug/ml) condition A condition B condition C 100 1924 1845 1233 2486 5118 8821 33.3333 1677 2383 2900 1914 8294 8104 11.1111 2833 2954 996 2597 7352 4725 3.7037 2824 3255 3059 3424 9414 8570 1.2346 3584 5974 5542 4382 6131 6853 0.4115 5517 5574 4724 3648 9027 11381 0.1372 5745 7826 7670 6880 9444 10124 0.0457 7498 7299 6703 9954 16101 13757 0.0046 22435 18765 26860 23215 13903 13795 0.0046 17613 22306 23633 21213 14095 10535

In an effort to determine if the peptides of the invention inhibited HCVpp entry at attachment or post-attachment steps, the peptide (sEQ ID NO: 77) was pre-incubated with cells at 4° C. followed by incubation with HCV pseudoparticles at 4° C. The entry process was allowed to complete after the cells were washed and incubated at 37° C. for 72 hours. If the peptide blocked or interfered with the attachment, few HCVpp particles should bind to the cells and lead to reduced luciferase signal. The peptide had approximately 50% inhibition whereas the anti-CD81 antibody inhibited entry by 20%. Alternatively, an explanation could be that the cell-associated peptide or antibody had no effects on virus attachment, but rather inhibit post-attachment steps, e.g. fusion. When we allowed the HCV pseudoparticles to attach first at 4° C., a nonpermissive temperature for fusion, and added the peptide or anti-CD81 antibody when post-attachment events occurred, anti-CD81 antibody almost inhibited entry by 100%, whereas, the peptide inhibited by approximately 80%. Our observation that anti-CD81 antibody inhibited HCVpp entry at a post-attachment step is consistent with published results (Cormier et al, Proc. Natl. Acad. Sci. 101:7270-7274). Our data also indicates that the peptide largely inhibits a post-attachment step.

The incubation scheme for the fusion/attachment experiments is summarized as follows:

TABLE 23 Data generated in fusion/attachment experiments average RLU V-2b (SEQ ID NO: 77) (peptide) anti-CD81 antibody A + P 527 1541 520 863 13 10 A 5196 7987 4644 5942 10851 6692 P 1984 2412 545 1647 13 15 Noinh. 13359 14364 17778 15167 13164 13994 average % control % control anti- anti-CD81 antibody (antibody) peptide CD81 antibody A + P 12 12 6 0 A 9662 9068 39 82 P 714 247 11 0 Noinh. 14957 14038 100 100 A = attachment; P = post attachment; Noinh = no inhibition

The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims.

Patents, patent applications, publications, product descriptions, and protocols are cited throughout this application, the disclosures of which are incorporated herein by reference in their entireties for all purposes.

Claims

1. An isolated polypeptide:

(i) comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 82-84, 87-97, 99, 101-103 and 117-138; or
(ii) consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3-81, 85, 86, 98, 100, 104-116 and 139.

2. The isolated polypeptide of claim 1:

(i) comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 82-84, 87-97, 99, 101-103, 117-130 and 132-138; or
(ii) consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 3, 7, 9, 10, 12-28, 30-38, 40, 42, 44-60, 62-68, 70-81, 85, 86, 98, 100 and 106-116.

3. A pharmaceutical composition comprising the polypeptide of claim 1 and a pharmaceutically acceptable carrier.

4. A composition comprising the polypeptide of claim 1 in association with one or more members selected from the group consisting of anti-human CD81 antibody, ribavirin, interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, and pegylated consensus interferon.

5. A kit comprising a polypeptide of claim 1 and a member selected from the group consisting of anti-human CD81 antibody, ribavirin, interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, and pegylated consensus interferon.

6. An isolated polynucleotide encoding the polypeptide of claim 1.

7. A recombinant vector comprising the polynucleotide of claim 6.

8. An isolated host cell comprising the vector of claim 7.

9. A method for making a polypeptide comprising culturing a host cell of claim 8 under conditions in which the polynucleotide is expressed.

10. The method of claim 9 wherein the polypeptide is isolated from the culture.

11. A method for inhibiting entry of a virus which is a member of the Flaviviridae family into a cell comprising contacting the cell with a polypeptide of claim 1.

12. The method of claim 11 wherein the cell is in vitro.

13. A method for treating or preventing infection of a subject with a virus which is a member of the Flaviviridae family comprising administering to said subject a therapeutically effective amount of a polypeptide of claim 1.

14. The method of claim 13 wherein the subject is a human.

15. The method of claim 13 wherein the virus is hepatitis C virus.

16. The method of claim 13 wherein the polypeptide is administered to the subject parenterally.

17. The method of claim 16 wherein the polypeptide is administered to the subject intramuscularly, intravenously or subcutaneously.

18. The method of claim 13 wherein said polypeptide is administered in association with one or more members selected from the group consisting of anti-human CD81 antibody, ribavirin, interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, and pegylated consensus interferon.

19. The method of claim 15 wherein said polypeptide is administered for a treatment time period sufficient to eradicate detectable hepatitic C virus-RNA and to maintain no detectable hepatitic C virus RNA for at least twelve weeks after the end of the treatment time period.

20. The method of claim 15 wherein said polypeptide is administered in association with a therapeutically effective amount of an interferon for a treatment time period sufficient to eradicate detectable hepatitic C virus-RNA and to maintain no detectable hepatitic C virus RNA for at least twelve weeks after the end of the treatment time period.

21. The method of claim 15, wherein the host is infected with multiple hepatitis C virus genotypes.

22. The method of claim 21, wherein the host is infected with hepatitis C virus genotype 1 hepatitis C virus genotype 2 or hepatitis C virus genotype 3.

23. The method of claim 13 wherein the subject is co-infected with human immunodeficiency virus (HIV).

24. A method for treating or preventing infection of a subject, with a virus which is a member of the Flaviviridae family of viruses, following transplantation of a liver into said host or transfusion of blood into said subject comprising administering to said subject a therapeutically effective amount of a polypeptide of claim 1.

25. The method claim 24 wherein the virus is hepatitis C virus.

26. The method of claim 24 wherein the polypeptide is administered in association with a member selected from the group consisting of anti-human CD81 antibody, ribavirin, interferon alfa-2a, interferon alfa-2b, interferon alfa-2c, interferon alfa n-1, interferon alfa n-3, consensus interferon, pegylated interferon alfa-2a, pegylated interferon alfa-2b, pegylated interferon alfa-2c, pegylated interferon alfa n-1, pegylated interferon alfa n-3, and pegylated consensus interferon.

27. The method of claim 24 wherein the polypeptide is administered to the subject parenterally.

28. The method of claim 27 wherein the polypeptide is administered to the subject intramuscularly, intravenously or subcutaneously.

Patent History
Publication number: 20100119483
Type: Application
Filed: Jul 23, 2009
Publication Date: May 13, 2010
Applicant:
Inventors: Rong Liu (Scotch Plains, NJ), Rumin Zhang (Edison, NJ), Rong Kong (Scotch Plains, NJ)
Application Number: 12/508,095