WO2010047829A1 - Polypeptides e2 mutants du virus de l'hépatite c pour le traitement du virus de l'hépatite c - Google Patents

Polypeptides e2 mutants du virus de l'hépatite c pour le traitement du virus de l'hépatite c Download PDF

Info

Publication number
WO2010047829A1
WO2010047829A1 PCT/US2009/005785 US2009005785W WO2010047829A1 WO 2010047829 A1 WO2010047829 A1 WO 2010047829A1 US 2009005785 W US2009005785 W US 2009005785W WO 2010047829 A1 WO2010047829 A1 WO 2010047829A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
mutant
antibody
virus
hcv
Prior art date
Application number
PCT/US2009/005785
Other languages
English (en)
Inventor
Mansun Law
Dennis R. Burton
Original Assignee
The Scripps Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Scripps Research Institute filed Critical The Scripps Research Institute
Publication of WO2010047829A1 publication Critical patent/WO2010047829A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/576Immunoassay; Biospecific binding assay; Materials therefor for hepatitis
    • G01N33/5767Immunoassay; Biospecific binding assay; Materials therefor for hepatitis non-A, non-B hepatitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/18Togaviridae; Flaviviridae
    • G01N2333/183Flaviviridae, e.g. pestivirus, mucosal disease virus, bovine viral diarrhoea virus, classical swine fever virus (hog cholera virus) or border disease virus
    • G01N2333/186Hepatitis C; Hepatitis NANB
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2469/00Immunoassays for the detection of microorganisms
    • G01N2469/20Detection of antibodies in sample from host which are directed against antigens from microorganisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • mutant hepatitis C virus (HCV) E2 polypeptides and nucleic acid molecules encoding the polypeptides are mutant hepatitis C virus (HCV) E2 polypeptides and nucleic acid molecules encoding the polypeptides.
  • the mutant E2 polypeptides and encoding nucleic acid molecules can be used for diagnosis and therapy of hepatitis C virus (HCV) infection and can be employed as vaccines for the prevention and treatment of HCV infection.
  • the mutant E2 polypeptides also can be employed for the production of antibodies against HCV.
  • HCV infection predisposes the patient to chronic liver cirrhosis, cancer and liver failure. About 85 % of individuals initially infected with HCV become chronically infected. Once established, chronic HCV infection causes an inflammation of the liver, and this can progress to scarring and eventually, liver cirrhosis. Some patients with cirrhosis will go on to develop liver failure or liver cancer. In the United States and Western Europe, the complications of chronic hepatitis and cirrhosis are the most common reasons for liver transplantation. In addition, liver disease caused by HCV is the leading cause of death in patients co-infected with human immunodeficiency virus. Given the large number of infected people worldwide, HCV infection can be a burden on health care systems worldwide.
  • mutant E2 polypeptides Provided herein are mutant E2 polypeptides, compositions containing mutant
  • the polypeptides can be employed for the prophylaxis and therapy of hepatitis C virus infection.
  • the polypeptides can be used as immunogens to elicit antibodies that can protect against infection by a hepatitis C virus (HCV).
  • HCV hepatitis C virus
  • the mutant HCV E2 polypeptides provided herein display conserved neutralizing AR3 epitopes recognized by conformation-dependent cross-neutralizing anti-HCV antibodies.
  • nucleic acid molecules encoding mutant HCV E2 polypeptides and expression vectors containing the nucleic acid molecules for their production.
  • cells that contain nucleic acid molecules encoding mutant HCV E2 polypeptides and cells that contain expression vectors containing the nucleic acid molecules.
  • preparations and pharmaceutical compositions containing a mutant HCV E2 polypeptide are also provided herein.
  • methods of eliciting an immune response in a mammal comprising administering a mutant HCV E2 polypeptide provided herein, methods for determining whether a mammal has been infected with an HCV, and methods for identifying an anti-HCV agent.
  • a mutant HCV E2 polypeptide contains the amino acid sequence which includes, from the amino to the carboxy termini: (1) a first segment that corresponds to amino acid residues 412 to 459 of a select HCV, (2) a second segment that corresponds to amino acid residues 486 to 569 of the select HCV, and (3) a third segment that corresponds to amino acid residues 581 to 645 of the select HCV.
  • the segments of the mutant HCV E2 polypeptide can be linked directly or via a linker, typically a polypeptide linker.
  • mutant HCV E2 polypeptides provided herein contain deletions of amino acid residues corresponding to amino acids 384 to 411 and amino acids 718 to 746 of the fiill-length E2 polypeptide of a select hepatitis C virus and does not include a contiguous sequence of amino acids that corresponds to amino acid residues 411 to 662 of the E2 polypeptide of the select hepatitis C virus.
  • mutant E2 polypeptides provided herein are polypeptides that can specifically or selectively bind to an antibody that is immunospecific for a conformational epitope on the E2 polypeptide that contains at least amino acids 411 to 462.
  • the conformational epitope contains amino acids corresponding to amino acids 412 to 424, 436 to 447 and 523 to 540 of the select hepatitis C virus.
  • the mutant E2 polypeptides provided herein contain (1) a first segment that corresponds to amino acid residues 412 to 459 of the E2 polypeptide of a select hepatitis C virus, (2) a second segment that corresponds to amino acid residues 486 to 569 of the E2 polypeptide of the select hepatitis C virus, and (3) a third segment that corresponds to amino acid residues 581 to 645 of the E2 polypeptide of the select hepatitis C virus, where the segments are linked directly or via a polypeptide linker, with the proviso that the mutant polypeptide does not include a contiguous sequence of amino acids corresponding to all of amino acid residues 411 to 662 of the E2 polypeptide of the select hepatitis C virus; the mutant polypeptide has deletions of amino acid residues corresponding to amino acids 384 to 411 and amino acids 718 to 746 relative to the full-length E2 polypeptide of the select hepatitis C virus.
  • the mutant E2 polypeptides provided herein do not contain the contiguous amino acid residues that correspond to amino acid residues 460 to 485 and/or 570 to 580 of the E2 polypeptide of the select hepatitis C virus.
  • the segments of the mutant E2 polypeptides provided herein can be linked directly or via a linker, such as a polypeptide linker provided that the polypeptide properly folds to form the conserved HCV E2 conformational epitope.
  • the linker located between the first and second segments contains 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or more amino acid residues provided that the polypeptide properly folds to form the conserved HCV E2 conformational epitope.
  • the linker located between the second and third segments contains 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or more amino acid residues provided that the polypeptide properly folds to form the conserved HCV E2 conformational epitope.
  • the linker located between the first and second segments contains about 26 amino acid residues. In some examples, the linker located between the second and third segments contains about 12 amino acid residues. In some examples, the first and second segments are linked via a linker that comprises amino acids corresponding to amino acids 460 to 485 of the full-length E2 polypeptide of a select hepatitis C virus. In some examples, the second and third segments are linked via a linker that comprises amino acids corresponding to amino acids 570 to 580 of the full-length E2 polypeptide of a select hepatitis C virus.
  • the segments of the mutant E2 polypeptide correspond to amino acid segments in E2 polypeptides in select hepatitis C virus such as subtypes Ia, Ib, Ic, 2a, 2b, 2c, 2i, 2k, 3a, 3b, 3k, 4a, 4d, 4f, 5a, 6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 61, 6m, 6n, 6o, 6q, 6p or 6t.
  • the select hepatitis C virus is a subtype 1 a, Ib, or 1 c virus.
  • the select hepatitis C virus is H77, HCV-L2, or HCV-G9.
  • the first segment of the mutant E2 polypeptide provide herein is 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %,
  • the second segment of the mutant E2 polypeptide provide herein is 65 %
  • the third segment of the mutant E2 polypeptide provided herein is 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % identical to amino acids 581 to 645 of hepatitis C virus H77.
  • the first amino acid segment of the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 888-912 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity therewith.
  • the second amino acid segment of the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS : 913-937 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity therewith.
  • the third amino acid segment of the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 938-962 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity therewith.
  • the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 727-730 and 740-742 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity therewith.
  • the mutant E2 polypeptide further includes a polypeptide tag, such as amino or carboxy terminal tag.
  • the tag is an N- terminal ubiquitin signal, a poly-histidine sequence, a FLAG (DYKDDDDK) sequence(SEQ ID NO:607), an HA sequence, a myc sequence, a V5 sequence, a chitin binding protein sequence, a maltose binding protein sequence or a glutathione- S-transferase sequence.
  • nucleic acid molecules that encode a mutant E2 polypeptide provided herein.
  • expression vectors that contain a nucleic acid molecule sequence that encodes a mutant E2 polypeptide provided herein.
  • the nucleic acid molecule encoding a polypeptide provided herein is operably linked to an expression control sequence or regulatory sequence.
  • the expression control sequence or regulatory sequence is a promoter.
  • the promoter is a viral promoter, a bacterial promoter or a mammalian promoter.
  • the promoter is a SV40 promoter, a Rous Sarcoma Virus promoter, or a cytomegalovirus immediate early promoter.
  • cells that contain a mutant E2 polypeptide provided herein are also provided herein. Also provided herein are cells that contain a nucleic acid molecule encoding a mutant E2 polypeptide provided herein. Also provided herein are cells that contain an expression vector that contains a nucleic acid molecule sequence encoding a mutant E2 polypeptide provided herein.
  • the cell is a bacterial cell, a mammalian cell, such as, for example, a Chinese hamster ovary cell.
  • the subject is a mammal, such as, but not limited to, mouse, sheep, goat, horse, rabbit, hamster, rat or human.
  • the subject is a human.
  • the method also involves obtaining a sample from the mammal, such as a blood, lymph, or urine sample.
  • the method includes administering to the subject a second dose of the polypeptide at a selected time after the first administration.
  • the methods provided herein further can involve isolating an antibody or antibody-producing cell from the mammal.
  • the antibody is a cross-neutralizing antibody.
  • the antibody is a conformation- dependent antibody.
  • the method further includes fusing the antibody-producing cells from the mammal with a myeloma cell to obtain an antibody-producing hybridoma.
  • the polypeptide is in a pharmaceutical composition that includes a pharmaceutically acceptable carrier. In some examples, the polypeptide is in an amount effective to prevent or treat HCV infection in the mammal.
  • the antibody can be a full length antibody (e.g. an IgG) or an antibody fragment (e.g. a Fab or F(ab')2).
  • the antibody is a monoclonal antibody, a single chain variable fragment (scFv), or other antigen binding fragment.
  • the antibody is a murine antibody.
  • the antibody is a conformation dependent antibody.
  • the antibody is a cross- neutralizing antibody.
  • the method involves administering to the mammal an expression vector that includes a nucleic acid molecule sequence encoding a polypeptide provided herein.
  • the nucleic acid molecule can include a sequence encoding a polypeptide of SEQ ID NO: 866, 867, 868, 869 or 870.
  • the nucleic acid molecule sequence can include the sequence of SEQ ID NO: 874, 875, 876, 877, 878, 879, 880 or 881.
  • the nucleic acid molecule is operably linked to an expression control sequence.
  • the expression control sequence can be a viral, phage, bacterial, or mammalian promoter.
  • the promoter can be a SV40 promoter, a Rous Sarcoma Virus promoter, or a cytomegalovirus immediate early promoter.
  • pharmaceutical compositions that include (1) a mutant E2 polypeptide provided herein, (2) a nucleic acid molecule that encodes a mutant E2 polypeptide provided herein, (3) a vector, such as an expression vector, that includes a nucleic acid molecule sequence encoding a mutant E2 polypeptide provided herein, or (4) an antibody that binds to a mutant E2 polypeptide provided herein, and a pharmaceutically acceptable carrier.
  • immunostimulatory or immunogenic compositions and vaccines that include a mutant E2 polypeptide provided herein or a nucleic acid molecule that encodes a mutant E2 polypeptide provided herein.
  • uses of a mutant E2 polypeptide provided herein or a nucleic acid molecule that encodes a mutant E2 polypeptide for the preparation of a pharmaceutical composition for the treatment or prevention of HCV infection in a subject are examples of a mutant E2 polypeptide provided herein or a nucleic acid molecule that encodes a mutant E2 polypeptide for the preparation of a pharmaceutical composition for the treatment or prevention of HCV infection in a subject.
  • purified preparations of the polypeptides provided herein in which at least 80 % of the polypeptides are in a conformation capable of binding to a conformation-dependent cross-neutralizing antibody.
  • purified preparations of the antibody provided herein in which the antibody is at least 5 % of the antibodies in the preparation are provided herein.
  • the sample can be a cell lysate, for example, a cell lysate from a bacterial, yeast, insect, or mammalian cell.
  • the method also involves separating the polypeptide from the antibody to obtain a preparation that has at least 50 % polypeptides.
  • the polypeptide is separated from the antibody by elution with 0.2 M glycine at pH 2.2; 2M sodium thiocyanate at pH 7.4; or 0.2 M glycine at pH 11.5.
  • the method further involves purifying the polypeptide using size- exclusion chromatography.
  • preparations obtained by a method provided herein in which at least 50 %, at least 75 %, at least 85 %, at least 90 %, or at least 95 % of the polypeptides in the preparation are in a conformation capable of binding to a conformation dependent antibody.
  • the conformation-dependent antibody binds specifically with a conformational epitope that includes: (1) amino acids 396-424 having the sequence TAGLVGLLTPGAKQNIQLINTNGSWHINS (SEQ ID NO: 694); (2) amino acids 436-447 having the sequence GWLAGLFYQHKF (SEQ ID NO: 695), and (3) amino acids 523-540 having the sequence GAPTYSWGANDTDVFVLN (SEQ ID NO: 696).
  • the conformation-dependent antibody binds specifically with a conformational epitope that includes: (1) amino acids 412-424 having the sequence QLINTNGSWHINS (SEQ ID NO: 688); (2) amino acids 436-447 having the sequence GWLAGLFYQHBCF (SEQ ID NO: 695), and (3) amino acids 523-540 having the sequence GAPTYSWGANDTDVFVLN (SEQ ID NO: 696).
  • the conformation-dependent antibody binds specifically with a conformational epitope on the HCV E2 polypeptide.
  • determining whether a mammal has been infected with a HCV involves contacting a sample, such as for example, a blood sample, from the mammal with a polypeptide provided herein and determining whether the polypeptide binds specifically with an antibody from the sample (e.g., a blood sample) of the mammal to form a polypeptide-antibody complex, wherein the presence of the complex indicates that the mammal has been infected with an HCV and the absence of the complex indicates that the mammal has not been infected with the virus.
  • a sample such as for example, a blood sample
  • identifying an anti-HCV agent that involves contacting a candidate agent with the E2 polypeptide provided herein, identifying the candidate agent as an anti-HCV agent if the candidate agent binds to the E2 polypeptide and prevents its binding with a cell receptor such as CD81 or prevents its ability to inhibit viral replication.
  • the purified polypeptides are useful as immunogens for raising broadly neutralizing antibodies against HCV, as reagents for diagnosis of HCV infection and for screening of new anti-HCV antiviral agents.
  • the polypeptides and corresponding coding nucleic acid molecules can be used as polypeptide- and nucleic acid-based vaccines to elicit a protective immune response against HCV.
  • methods of immunizing a mammal against HCV that involves administering a polypeptide- or nucleic acid-based vaccine.
  • FIGS IA-C illustrate properties of anti-HCV E2 Fabs isolated as described herein.
  • FIG. IA illustrates the specificity of anti-E2 Fabs. The binding of Fabs to GST-E1E2 complex and E2 is compared. The GST-E 1E2 fusion protein was captured by a goat anti-GST antibody while soluble E2 and ovalbumin were coated directly onto ELISA plates. Fabs were added to the antigens and subsequently detected with phosphatase-conjugated goat anti-human F(ab)'2 IgG. Recombinant Fabs were produced in cleared lysate of E. coli transformed with the corresponding phagemids.
  • FIG. IA illustrates the specificity of anti-E2 Fabs. The binding of Fabs to GST-E1E2 complex and E2 is compared. The GST-E 1E2 fusion protein was captured by a goat anti-GST antibody while soluble E2 and ovalbumin were coated directly onto ELISA plates
  • IB illustrates competition between MAb AR3A and anti-E2 Fabs.
  • Vaccinia-expressed El E2 was captured onto ELISA wells by lectin and preincubated with saturating concentration of soluble Fabs before the addition of MAb AR3A.
  • Binding of MAb AR3A was detected with a goat anti-human IgG Fc antibody and the % reduction of binding compared to that in the absence of a Fab is shown. Lightly-shaded bars indicate that Fabs bind E2 better than El E2; while bars of medium shading indicate that Fabs bind E1E2 better than E2.
  • FIG. 1C illustrates the inhibition of anti-E2 Fab binding to El E2 by mouse MAb H53.
  • FIG. 2 shows neutralization of HCVpp by human Fabs. Infectivity in Relative
  • RLU Light Units
  • MMV murine leukemia virus
  • GT Ia H77
  • GT Ib H77
  • GT Ib OH8
  • GT Ib GT Ib
  • FIG. 2E J6 (GT 2a)
  • ARl-Fabs B2, Dl & E
  • AR2-Fabs F & G
  • AR3- Fabs Cl, J2, J3 & L4.
  • Control anti-HIV gpl20 Fab bl2; Empty, background infectivity from pseudotype virus generated without Env gene.
  • Dotted lines indicate HCVpp infectivity in the absence of any antibody. Error bars represent SEM calculated from three experiments performed in the same manner.
  • FIG. 3 is a schematic diagram of E2 regions important for binding of AR3- specific antibodies.
  • E2 (residues 384-746) is a transmembrane glycoprotein, and a truncated form of E2 (residues 384-661) can be expressed as a soluble protein that retains its ability to bind cell lines expressing HCV receptors and CD81 -LEL (Michalak, J.P. et al. J. Gen. Virol. 78, 2299-2306 (1997)).
  • the regions that were investigated by antibody competition and alanine mutagenesis are indicated by dotted and solid boxes, respectively.
  • the AR3 discontinuous epitopes include: (1) amino acids 396-424 having the sequence TAGLVGLLTPGAKQNIQLINTNGSWHINS (SEQ ID NO: 694); (2) amino acids 436-447 having the sequence GWLAGLFYQHKF (SEQ ID NO: 695), and (3) amino acids 523-540 having the sequence GAPTY S WGANDTD VF VLN (SEQ ID NO: 696). Important residues in these regions include S424, G523, P525, G53O, D535, V538 and N540. The locations of the N-linked glycans are indicted by branched forks. The hypervariable regions (see Troesch, M. et al. Virology 352, 357-367 (2006) and the transmembrane regions are indicated by the designation HVRs and TM.
  • FIG. 4 illustrates the levels of human MAb in human liver-chimeric mice 24 hours post-administration.
  • Intravenous administration of human serum is the most reliable way to assure delivery of human serum but a stressful procedure: 5 of 18 treated mice did not recover after the procedure.
  • Human IgG in mouse sera were quantified by a quantitative sandwich ELISA using conjugated and unconjugated goat anti-human F(ab)' 2 antibody.
  • FIG. 5 demonstrates passive antibody protection against an HCV population.
  • FIG. 6 is a sequence comparison of a viral quasispecies population in the HCV genotype la-infected human serum.
  • Partial E2 amino acid sequences (residues 384-622) of a total of 40 clones represented by KP S9 (SEQ ID NO: 701), KP R14 (SEQ ID NO: 702), KP S6 (SEQ ID NO: 703), KP S 18 (SEQ ID NO: 704), KP S 16 (SEQ ID NO: 705), KP R8 (SEQ ID NO: 706), KP S20 (SEQ ID NO: 707), KP S4 (SEQ ID NO: 708), KP R3 (SEQ ID NO: 709), KP S3 (SEQ ID NO: 710), KP S 12 (SEQ ID NO: 711), KP S15 (SEQ ID NO: 712), KP S5 (SEQ ID NO: 713), KP R7 (SEQ ID NO: 714), KP Rl 1 (SEQ ID NO: 715), KP Rl (SEQ ID NO: 716), KP R12 (SEQ ID NO: 717), KP
  • the top sequence, clone KP S9 represents the consensus and dominant sequence in this infectious serum.
  • the periods indicate regions of amino acid sequence identity.
  • the frequency of each clone is bracketed.
  • Hypervariable regions (HVRs) are within the dashed-line boxes. Regions important for binding of AR3-antibodies are within the solid-line boxes.
  • the corresponding sequences of isolates H77 (SEQ ID NO: 723) and UKNlbl2.16 (SEQ ID NO: 724), sharing 87% and 75% amino acid identity with KP S9, respectively, are shown for comparison.
  • FIG. 7 is a schematic illustration of a panel of recombinant E2 fragments.
  • Full length E2 (residues 384-746) is shown at the top and the relative locations of N- gl yeans and cysteines are marked by light and darker vertical lines, respectively.
  • the hypervariable region 1 (HVRl) at the N-terminus and transmembrane region at the C- terminus of E2 are shaded.
  • the positions of N- or C-terminal truncation in the mutants are indicated, and the Flag tags are indicated by boxes at the C-termini. Fragments are named according to the primer sets used in gene amplification.
  • disulfide bridges are predicted to form between C1-C16 (i.e. residues C429-C644), C2-C4 (C452-C486), C8-C9 (C552-C564), C13-C14 (C597-C607), and C7-C11 (C508-581) or C7-C12 (C581-585).
  • FIG. 8A-H illustrate the binding properties of E2 fragments.
  • 293T cells were transfected with DNA plasmids encoding the E2 fragments depicted in FIG. 7 and the expression of the corresponding proteins was assayed by sandwich ELISA.
  • ELISA wells were pre-coated with MAbs specific to the 3 different E2 antigenic regions (ARl, AR2 and AR3), or CD81-LEL, to capture the recombinant proteins in serially diluted cell supernatants. The reagents used in the capture are indicated on the left of the bar charts.
  • Bound E2 fragments were detected using the mouse anti-Flag tag M2 MAb (Sigma). Data shown are means of duplicate measurements.
  • E2flr2a produced in the presence of kifunensine was purified using a MAb AR3A- affinity column and was eluted with low pH (0.2 M glycine pH 2.2), 2 M NaSCN (pH 7.4) or high pH (0.2 M glycine pH 11.5) buffer.
  • the different purified E2flr2a monomers were titrated from 4 ⁇ g/mL ( ⁇ 145 nM, 5-fold serial dilution) to investigate their antigenicities.
  • the purified proteins were captured onto microwells precoated with Galanthus nivalis lectins (5 ⁇ g/mL) and the captured proteins detected with the indicated human anti-E2 monoclonal antibodies (MAbs).
  • MAbs human anti-E2 monoclonal antibodies
  • CD81-LEL microwells coated with maltose binding protein (MBP)-fused CD81-large extracellular loop (LEL) (10 ⁇ g/mL) were used to capture the purified proteins and bound proteins were detected with the mouse anti-FLAG tag MAb M2.
  • MBP maltose binding protein
  • LEL CD81-large extracellular loop
  • Bound human or mouse MAbs were detected with peroxidase-conjugated anti-human or anti-mouse secondary antibodies and TMB substrate. The results show that E2flr2a monomers eluted by buffers with different pH are similar antigenically.
  • FIG. 10A-E are graphs illustrating the antigenic properties of E2 ⁇ TM.
  • E2 ⁇ TM produced in the presence of kifunensine was purified using a MAb AR3A- affinity column and was eluted with 2 M NaSCN buffer (pH 7.4). The effect of pH on the antigenicity of the protein was investigated. Purified E2 ⁇ TM monomers were exposed briefly to low or high pH by adding 10-fold excess volume of 0.2M glycine pH 2.2 or pH 11.5, respectively. After 10 minutes, the solutions were neutralized by adding equal volume of 2M Tris-HCl pH 7.4 and treated and untreated E2 ⁇ TM monomers were titrated from 5.5 ⁇ g/mL ( ⁇ 145 nM, 5-fold serial dilution).
  • the purified proteins were captured onto microwells precoated with Galanthus nivalis lectins (5 ⁇ g/mL) and the captured proteins detected with the indicated human anti-E2 monoclonal antibodies (MAbs).
  • MAbs human anti-E2 monoclonal antibodies
  • CD81-LEL microwells coated with maltose binding protein (MBP)-fused CD81 -large extracellular loop (LEL) (10 ⁇ g/mL) were used to capture the purified proteins and bound proteins were detected with the mouse anti-FLAG tag MAb M2.
  • Bound human or mouse MAbs were detected with peroxidase-conjugated anti-human or anti-mouse secondary antibodies and TMB substrate. The results show that pH does not have a significant effect on the antigenicity of E2 ⁇ TM. DETAILED DESCRIPTION
  • hepatitis C virus includes different viral genotypes, subtypes, quasispecies and isolates. It includes any noncytopathic RNA virus that has a single and positive-stranded RNA genome belonging to the Hepacivirus genus of the Flaviviridae family.
  • the term includes different isolates of HCV such as, without limitation, those having polyprotein sequences and accession numbers shown above, as well as any others in the NCBI database. Examples of different genotypes encompassed by this term include, without limitation, genotype 1, 2, 3, 4, 5 and 6, as described in Simmonds et al. (Hepatology 42:962-973). Reference to HCV also includes those of any additional genotypes that are established.
  • Examples of different subtypes include, without limitation, Ia, Ib, Ic, 2a, 2b, 2c, 2i, 2k, 3a, 3b, 3k, 4a, 4d, 4f, 5a, 6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 61, 6m, 6n, 6o, 6q, 6p and 6t .
  • the term also includes cell culture HCVs (HCVcc) and pseudotype HCVs (HCVpp), as well as HCV quasispecies. Various HCVs are described by Simmonds P.
  • HCV nucleotide sequences are known in the art. For example, see the Viral Bioinfomatics Research Center (hcvdb.org) and the Hepatitis C Virus database (hcv.lanl.gov/).
  • An "E2" polypeptide is the HCV viral envelope protein that forms a heterodimer with the El glycoprotein through non-covalent interactions.
  • HCV El and E2 envelope glycoproteins are exposed on the viral surface where they function in viral attachment and fusion to target cells.
  • the E2 glycoprotein is residues 384 to 746.
  • the term "mutant" as used in reference to an HCV E2 polypeptide provided herein means that the polypeptide is modified by one or more substitutions or deletions relative to a naturally occurring HCV E2 polypeptide.
  • the mutant polypeptide provided herein is free of sequences in the hypervariable region of the HCV E2 polypeptide, in particular, sequences that correspond to the segment defined by amino acid residues 384 to 395.
  • mutant E2 polypeptides provided herein that contain one or more deletions relative to a naturally occurring E2 are able to fold into a conformation that preserves a conformational epitope, such as for example, a conformational epitope recognized by an AR3A, AR3B, AR3C, or AR3D antibody.
  • Exemplary mutant E2 polypeptides provided herein differ from the corresponding naturally-occurring E2 amino acid sequence in that the mutant E2 polypeptide provided herein does not include one or more segments defined by (1) amino acid residues 384 to 411; (2) amino acid residues 460 to 485; (3) amino acid residues 570-580, (4) amino acid residues 646-661, (5) amino acid residues 662-717 or 718-746, or any combination thereof, relative to the naturally-occurring E2 polypeptide.
  • the mutant E2 polypeptides provided herein also can have one or more amino acid modifications that decrease the immunogenicity of particular immunodominant epitopes, such as for example epitopes recognized by the ARIA and ARlB antibodies.
  • Such modifications include, but are not limited to deletion or substitution of amino acids at positions 416, 417, 483, 484, 485, 538, 540, 544, 545, 547, 549 or any combinations thereof relative to the E2 polypeptide sequence of HCV (e.g. HCV strain H77).
  • the recitation that a sequence of amino acids "corresponds to" particular amino acids from a selected isolate of HCV means the corresponding amino acids in another isolate. Since E2 and other proteins may include slight amino acid variations from isolate-to-isolate, the exact amino acid residues or positions may vary by one or two residues.
  • One of skill in the art readily can align, using standard programs, or by eye, since the amount of variation is minimal, polypeptides from different isolates to identify corresponding segments or residues.
  • a "modification" is in reference to modification of a sequence of amino acids of a polypeptide or a sequence of nucleotides in a nucleic acid molecule and includes deletions, insertions, and replacements of amino acids and nucleotides, respectively.
  • Methods for modifying a polypeptide are routine to those of skill in the art, such as by using recombinant DNA methodologies or direct synthesis.
  • deletion when referring to a nucleic acid molecule or polypeptide sequence, refers to the deletion of one or more nucleotides or amino acids compared to a sequence, such as a target polynucleotide or polypeptide or a native or wild-type sequence.
  • insertion when referring to a nucleic acid molecule or amino acid sequence, describes the inclusion of one or more additional nucleotides or amino acids, within a target, native, wild-type or other related sequence.
  • a nucleic acid molecule that contains one or more insertions compared to a wild-type sequence contains one or more additional nucleotides within the linear length of the sequence.
  • additions to nucleic acid and amino acid molecules describe addition of nucleotides or amino acids onto either termini compared to another nucleic acid or amino acid molecule.
  • substitution refers to the replacing of one or more nucleotides or amino acids in a native, target, wild-type or other nucleic acid molecule or polypeptide with an alternative nucleotide or amino acid, without changing the length (as described in numbers of residues) of the molecule.
  • substitutions in a molecule does not change the number of amino acid residues or nucleotides of the molecule.
  • Substitution mutations compared to a particular polypeptide can be expressed in terms of the number of the amino acid residue along the length of the polypeptide sequence.
  • a modified polypeptide having a modification in the amino acid at the 19 l position of the amino acid sequence that is a substitution of Isoleucine (He; I) for cysteine (Cys; C) can be expressed as Il 9C, Ilel9C, or simply C 19, to indicate that the amino acid at the modified 19 th position is a cysteine.
  • the molecule having the substitution has a modification at He 19 of the unmodified polypeptide.
  • polypeptide refers to a polymer of three or more amino acids, regardless of post-translational modifications such as methylation, glycosylation or phosphorylation.
  • a "peptide” refers to a polypeptide that is from 2 to about or 40 amino acids in length.
  • an “amino acid” is an organic compound containing an amino group and a carboxylic acid group.
  • a polypeptide contains two or more amino acids.
  • amino acids contained in the antibodies provided include the twenty naturally-occurring amino acids (Table 1), non-natural amino acids, and amino acid analogs (e.g., amino acids wherein the ⁇ -carbon has a side chain).
  • the amino acids, which occur in the various amino acid sequences of polypeptides appearing herein are identified according to their well-known, three- letter or one-letter abbreviations (see Table 1).
  • amino acid residue refers to an amino acid formed upon chemical digestion (hydrolysis) of a polypeptide at its peptide linkages.
  • the amino acid residues described herein are generally in the "L” isomeric form. Residues in the "D” isomeric form can be substituted for any L-amino acid residue, as long as the desired functional property is retained by the polypeptide.
  • NH2 refers to the free amino group present at the amino terminus of a polypeptide.
  • COOH refers to the free carboxy group present at the carboxyl terminus of a polypeptide.
  • amino acid residues represented herein by a formula have a left to right orientation in the conventional direction of amino-terminus to carboxyl- terminus.
  • amino acid residue is defined to include the amino acids listed in the Table of Correspondence (Table 1), modified, non-natural and unusual amino acids.
  • a dash at the beginning or end of an amino acid residue sequence indicates a peptide bond to a further sequence of one or more amino acid residues or to an amino-terminal group such as NH 2 or to a carboxyl-terminal group such as COOH.
  • Suitable conservative substitutions of amino acids are known to those of skill in this art and generally can be made without altering a biological activity of a resulting molecule.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al., Molecular Biology of the Gene, 4th Edition, 1987, The Benjamin/Cummings Pub. co., p.224).
  • naturally occurring amino acids refer to the 20 L-amino acids that occur in polypeptides.
  • non-natural amino acid refers to an organic compound that has a structure similar to a natural amino acid but has been modified structurally to mimic the structure and reactivity of a natural amino acid.
  • Non- naturally occurring amino acids thus include, for example, amino acids or analogs of amino acids other than the 20 naturally occurring amino acids and include, but are not limited to, the D-isostereomers of amino acids.
  • non-natural amino acids are known to those of skill in the art, and include, but are not limited to, 2- Aminoadipic acid (Aad), 3-Aminoadipic acid (Baad), ⁇ -alanine/ ⁇ -Amino-propionic acid (BaIa), 2-Aminobutyric acid (Abu), 4-Aminobutyric acid/piperidinic acid (4Abu), 6-Aminocaproic acid (Acp), 2-Aminoheptanoic acid (Ahe), 2-Aminoisobutyric acid (Aib), 3-Aminoisobutyric acid (Baib), 2-Aminopimelic acid (Apm), 2,4- Diaminobutyric acid (Dbu), Desmosine (Des), 2,2'-Diaminopimelic acid (Dpm), 2,3- Diaminopropionic acid (Dpr), N-Ethylglycine (EtGIy), N-Ethylasparagine (EtA
  • a "native polypeptide” or a “native nucleic acid” molecule is a polypeptide or nucleic acid molecule, respectively, that can be found in nature.
  • a native polypeptide or nucleic acid molecule can be the wild-type form of a polypeptide or nucleic acid molecule.
  • a native polypeptide or nucleic acid molecule can be the predominant form of the polypeptide, or any allelic or other natural variant thereof.
  • the variant polypeptides and nucleic acid molecules provided herein can have modifications compared to native polypeptides and nucleic acid molecules.
  • the wild-type form of a polypeptide or nucleic acid molecule is a form encoded by a gene or by a coding sequence encoded by the gene. Typically, a wild-type form of a gene, or molecule encoded thereby, does not contain mutations or other modifications that alter function or structure.
  • the term wild-type also encompasses forms with allelic variation as occurs among and between species and virus genotype and subtype variations.
  • a predominant form of a polypeptide or nucleic acid molecule refers to a form of the molecule that is the major form produced from a gene. A "predominant form" varies from source to source.
  • polypeptides for example, by alternative splicing and/or by alternative protein processing.
  • a different polypeptide can be a "predominant form.”
  • numeric terms identifying amino acid residues or positions in a polypeptide i.e. the protein or polypeptide "coordinates," for example, the term “residues 396 to 424," “residue 416,” or “amino acid 416,” are based on the absolute amino acid numbering system for HCV described by Kuiken et al. in Hepatology 44: 1355-1361 (2006), which is incorporated herein by reference in its entirety.
  • the polyprotein sequence of HCV strain WIl is used as a reference in the numbering system, and the first amino acid of the core protein is amino acid residue number 1.
  • Other HCV polyprotein sequences are compared with the H77 polyprotein sequences by sequence alignment. Insertions in other non-H77 sequences are identified using a residue number/alphabet designation relative to the H77 reference. For example, three inserted amino acids in a non-H77 polyprotein sequence inserted between amino acid residues 396 and 397 of the reference H77 sequence would be identified as follows: residue 396a, 396b and 396c.
  • Insertions longer than the length of the alphabet would be identified as follows: ...396x, 396y, 396z, 396aa, 396ab, 396ac, ... 396ax, 396ay, 396az, 396ba, 396bb ... .
  • Deletions in a non-H77 sequence relative to the H77 reference sequence can be indicated by identifying the residue deleted. For example, a missing residue, i.e. a "deletion”, in a non-H77 sequence relative to the H- 77 reference sequence identified in a sequence alignment such as a deletion of amino acid residue 396 is indicated by the term "del 396".
  • a polypeptide coordinate or coordinates such as “amino acid 396,” “residue 396,” or “amino acids 396 to 424,” refer to analogous residues or segments in HCV polyproteins from different isolates, strains, subtypes or genotypes. Analogous residues or segments can be identified by sequence alignment as described below. A similar system is used for identifying HCV nucleotide sequence.
  • amino acid sequences of two or more HCV E2 polypeptides can be compared by alignment using methods known in the art including but not limited to, those described in Computational Molecular Biology, Lesk, A. N., ed., Oxford University Press, New York (1988), Biocomputing: Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I, Griffin, A.M., and Griffin, H. G., eds., Humana Press, New Jersey (1994); Sequence Analysis in Molecular Biology, von Heinge, G., Academic Press (1987); Sequence Analysis Primer, Gribskov, M.
  • Two HCV polyprotein sequences can be compared by sequence alignment in a manner to produce the highest degree of sequence similarity or identity. Upon such alignment, sequence identity is determined on a position-by-position basis, e.g., the sequences are "identical" at a particular position if at that position, the amino acid residues are identical. Exemplary methods to determine sequence identity between two sequences are designed to give the largest match between the sequences tested. Methods to determine sequence identity are codified in publicly available computer programs.
  • Such programs include, but are not limited to, the GCG program package (Devereux, et ah, Nucleic Acids Research, 12:387 (1984)), BLASTP, BLASTN and FASTA (Altschul et al., J. Molec. Biol., 215:403 (1990)).
  • the BLASTX program is publicly available from NCBI and other sources (BLAST Manual, Altschul et ah, NCVI NLM NIH
  • HCV sequence analysis tools are known and available in the art, such as for example, but not limited to, online tools available from the Hepatitis C Virus (HCV) Database Project provided by the Los Alamos National laboratory (hcv.lanl.gov) and the European HepCVax Cluster provided by the Leiden University Medical Center, The Netherlands (euhcvdb.ibcp.fr).
  • HCV Hepatitis C Virus
  • amino acid 396 or “amino acids 396 to 424" refers to analogous residues in different HCVs including, for example, HCVs of different isolates, strains, species, quasispecies, subtypes or genotypes.
  • antibody refers to immunoglobulins and immunoglobulin fragments, whether natural or partially or wholly synthetically, such as recombinantly, produced, including any fragment thereof containing at least a portion of the variable region of the immunoglobulin molecule that retains the binding specificity ability of the full-length immunoglobulin.
  • an antibody includes any protein having a binding domain that is homologous or substantially homologous to an immunoglobulin antigen-binding domain (antibody combining site).
  • Antibodies include antibody fragments, such as anti-RSV antibody fragments.
  • the term antibody thus, includes synthetic antibodies, recombinantly produced antibodies, multispecific antibodies (e.g., bispecific antibodies), human antibodies, non-human antibodies, humanized antibodies, chimeric antibodies, intrabodies, and antibody fragments, such as, but not limited to, Fab fragments, Fab' fragments, F(ab') 2 fragments, Fv fragments, disulfide-linked Fvs (dsFv), Fd fragments, Fd' fragments, single-chain Fvs (scFv), single-chain Fabs (scFab), diabodies, anti-idiotypic (anti-Id) antibodies, or antigen-binding fragments of any of the above.
  • Fab fragments such as, but not limited to, Fab fragments, Fab' fragments, F(ab') 2 fragments, Fv fragments, disulfide-linked Fvs (dsFv), Fd fragments, Fd' fragments, single-chain Fvs (scFv), single-chain Fab
  • Antibodies provided herein include members of any immunoglobulin type (e.g., IgG, IgM, IgD, IgE, IgA and IgY), any class (e.g. IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass (e.g., IgG2a and IgG2b).
  • immunoglobulin type e.g., IgG, IgM, IgD, IgE, IgA and IgY
  • any class e.g. IgGl, IgG2, IgG3, IgG4, IgAl and IgA2
  • subclass e.g., IgG2a and IgG2b.
  • an “antibody fragment” refers to any portion of a full-length antibody that is less than full length but contains at least a portion of the variable region of the antibody that binds antigen (e.g. one or more CDRs and/or one or more antibody combining sites) and thus retains the binding specificity, and at least a portion of the specific binding ability of the full-length antibody; antibody fragments include antibody derivatives produced by enzymatic treatment of full-length antibodies, as well as synthetically, e.g. recombinantly produced derivatives. An antibody fragment is included among antibodies.
  • antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, single-chain Fvs (scFv), Fv, dsFv, diabody, Fd and Fd' fragments other fragments, including modified fragments (see, for example, Methods in Molecular Biology, VoI 207: Recombinant Antibodies for Cancer Therapy Methods and Protocols (2003); Chapter 1 ; p 3-25, Kipriyanov).
  • the fragment can include multiple chains linked together, such as by disulfide bridges and/or by peptide linkers.
  • An antibody fragment generally contains at least or about 50 amino acids and typically at least or about 200 amino acids.
  • an Ig domain is a domain, recognized as such by those in the art, that is distinguished by a structure, called the Immunoglobulin (Ig) fold, which contains two beta-pleated sheets, each containing anti-parallel beta strands of amino acids connected by loops. The two beta sheets in the Ig fold are sandwiched together by hydrophobic interactions and a conserved intra-chain disulfide bond.
  • Individual immunoglobulin domains within an antibody chain further can be distinguished based on function. For example, a light chain contains one variable region domain (VL) and one constant region domain (C L ), while a heavy chain contains one variable region domain (VH) and three or four constant region domains (C H ).
  • VL variable region domain
  • C L constant region domain
  • C H variable region domain
  • Each V L , C L , V H, and CH domain is an example of an immunoglobulin domain.
  • variable domain or variable region is a specific Ig domain of an antibody heavy or light chain that contains a sequence of amino acids that varies among different antibodies.
  • Each light chain and each heavy chain has one variable region domain, V L and V H , respectively.
  • the variable domains provide antigen specificity, and thus are responsible for antigen recognition.
  • Each variable region contains CDRs that are part of the antigen-binding site domain and framework regions (FRs).
  • antigen-binding domain As used herein, "antigen-binding domain,” “antigen-binding site,” “antigen combining site” and “antibody combining site” are used synonymously to refer to a domain within an antibody that recognizes and physically interacts with cognate antigen.
  • a native conventional full-length antibody molecule has two conventional antigen-binding sites, each containing portions of a heavy chain variable region and portions of a light chain variable region.
  • a conventional antigen-binding site contains the loops that connect the anti-parallel beta strands within the variable region domains.
  • the antigen combining sites can contain other portions of the variable region domains.
  • Each conventional antigen-binding site contains three hypervariable regions from the heavy chain and three hypervariable regions from the light chain. The hypervariable regions also are called complementarity-determining regions (CDRs).
  • framework regions are the domains within the antibody variable region domains that are located within the beta sheets; the FRs are comparatively more conserved, in terms of their amino acid sequences, than the hypervariable regions.
  • a "constant region” domain is a domain in an antibody heavy or light chain that contains a sequence of amino acids that is comparatively more conserved than that of the variable region domain.
  • each light chain has a single light chain constant region (C L ) domain and each heavy chain contains one or more heavy chain constant region (C H ) domains, which include, C H I, C H 2, C H 3 and C H 4.
  • C L constant region
  • C H heavy chain constant region
  • Full-length IgA, IgD and IgG isotypes contain CHU CH2, CH3 and a hinge region, while IgE and IgM contain C H I, C H 2, C H 3 and C H 4.
  • C H I and C L domains extend the Fab arm of the antibody molecule, thus contributing to the interaction with antigen and rotation of the antibody arms.
  • Antibody constant regions can serve effector functions, such as, but not limited to, clearance of antigens, pathogens and toxins to which the antibody specifically binds, e.g. through interactions with various cells, biomolecules and tissues.
  • the term "binds specifically” or “specifically binds,” with reference to an antibody/antigen interaction means that the antibody binds with a particular antigen without substantially binding to other unrelated antigens.
  • the antibody has at least 50 % or greater affinity, such as about 75 % or greater affinity, such as, for example, about 90 % or greater affinity, to a particular polypeptide than to other unrelated polypeptides.
  • an antibody that specifically binds (or that immunospecifically binds) to a virus antigen or virus is one that binds to a virus antigen (or to the antigen in the virus or to the virus) with an affinity constant (K 3 ) of about or IxIO 7 M '1 or Ix 10 8 M ' 'or greater (or a dissociation constant (K d ) of Ix 10 "7 M or IxIC 8 M or less).
  • Affinity and dissociation constants can be determined by standard kinetic methodology for antibody reactions, for example, immunoassays, surface plasmon resonance (SPR) (Rich and Myszka (2000) Curr. Opin. Biotechnol 11 :54; Englebienne (1998) Analyst. 123: 1599), isothermal titration calorimetry (ITC) or other kinetic interaction assays known in the art (see, e.g., Paul, ed., Fundamental Immunology, 2nd ed., Raven Press, New York, pages 332-336
  • neutralize means that the antibody can prevent or reduce HCV infection or replication in a cell culture or in a mammal, as well as alleviate one or more symptoms associated with HCV infection in a mammal.
  • reduce means a decrease in any amount such as a 5 %, 10 %, 15 %, 20 %, 25 %, 30 %, 35 %, 40 %, 45 %, 50 %, 55 %, 60 %, 65 % or more than 65 %.
  • HCV infection or replication can be detected by examining HCV RNA levels, virus particles count or clinical symptoms associated with HCV infection using methods known to those of skill in the art.
  • Whether an antibody will prevent or reduce HCV infection or replication or alleviate associated symptoms can be determined using methods known in the art, as well as the methods described herein, including determining the level of HCV RNA in a sample from a mammal that has been infected with HCV or detecting reduction of signals from a reporter gene encoded by the virus such as, for example, the relative light unit (RLU) for luciferase or the mean fluorescence intensity (MFI) of green fluorescent protein (GFP).
  • RLU relative light unit
  • MFI mean fluorescence intensity
  • an antibody will bind selectively to HCV and neutralize it can be determined using methods known in the art, as well as the methods described herein, including determining the level of HCV RNA or detecting reduction of signals from a reporter gene encoded by the virus such as, for example, the relative light unit (RLU) for luciferase or the mean fluorescence intensity (MFI) of green fluorescent protein (GFP).
  • RLU relative light unit
  • MFI mean fluorescence intensity
  • GFP green fluorescent protein
  • epitopic determinants typically comprise chemically active surface groupings of molecules such as amino acids or sugar side chains and typically have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • epitope refers to any antigenic determinant on an antigen to which the paratope of an antibody binds, and is made up of one or more segments of amino acids.
  • An epitope can be a linear or conformational epitope, and can be continuous or discontinuous. Typically, linear epitopes are continuous, i.e. made up of one continuous stretch of amino acids. Conformational epitopes can be discontinuous i.e. made up of two or more discontinuous segments of amino acids that come together to form an epitope when the antigen is folded. Methods for determining whether antibodies bind to the same epitope are known in the art. Epitopes can be defined or mapped by standard methods well known in art. For example, epitopes can be mapped using assays, such as ELISA assays, utilizing peptide libraries or site-directed mutagenesis of the antigen (such as alanine-scanning of the antigen).
  • binds to the same epitope with reference to two or more antibodies means that the antibodies compete for binding to an antigen and bind to the same, overlapping or encompassing continuous or discontinuous segments of amino acids.
  • Those of skill in the art understand that the phrase "binds to the same epitope” does not necessarily mean that the antibodies bind to exactly the same amino acids.
  • the precise amino acids to which the antibodies bind can differ.
  • a first antibody can bind to a segment of amino acids that is completely encompassed by the segment of amino acids bound by a second antibody.
  • a first antibody binds one or more segments of amino acids that significantly overlap the one or more segments bound by the second antibody.
  • such antibodies are considered to "bind to the same epitope.”
  • Antibody competition assays can be used to determine whether an antibody "binds to the same epitope" as another antibody. Such assays are well known in the art and are described herein (see, e.g. Examples 1-4). Typically, competition of 70 % or more, such as 70%, 71%, 72%, 73%, 74%, 75%, 80%, 85%, 90%, 95% or more, of an antibody known to interact with the epitope by a second antibody under conditions in which the second antibody is in excess and the first saturates all sites, is indicative that the antibodies "bind to the same epitope.” To assess the level of competition between two antibodies, for example, radioimmunoassays or assays using other labels for the antibodies, such as biotin (see, e.g., Example 1) can be used.
  • an HCV antigen such as the E1E2 complex
  • a saturating amount of a first anti-HCV antibody or antigen-binding fragment thereof conjugated to a labeled compound e.g., 3 H, 125 I or biotin
  • a labeled compound e.g. 3 H, 125 I or biotin
  • the amount of labeled antibody that is bound to the antigen in the presence of the unlabeled blocking antibody is then assessed and compared to binding in the absence of the unlabeled blocking antibody. Competition is determined by the percentage change in binding signals in the presence of the unlabeled blocking antibody compared to the absence of the blocking antibody.
  • first and second antibody of 70 % or more such as 70%, 71%, 72%, 73%, 74%, 75%, 80%, 85%, 90%, 95% or more
  • first antibody inhibits binding of the second antibody (or vice versa) to the antigen by 70%, 71%, 72%, 73%, 74%, 75%, 80%, 85%, 90%, 95% or more (compared to binding of the antigen by the second antibody in the absence of the first antibody).
  • inhibition of binding of a first antibody to an antigen by a second antibody of 70%, 71%, 72%, 73%, 74%, 75%, 80%, 85%, 90%, 95% or more indicates that the two antibodies bind to the same epitope.
  • the term "bind selectively” or “selectively binds,” in reference to a polypeptide or an antibody provided herein, means that the polypeptide or antibody binds with a selected epitope without substantially binding to another epitope.
  • an antibody or fragment thereof that selectively binds to a selected epitope specifically binds to the epitope, such as with an affinity constant (K a ) of about or IxIO 7 M “1 or 1 x 10 8 M-Or greater, as defined below.
  • telomere binding fragment As used herein, “specifically binds” or “immunospecifically binds,” with respect to an antibody or antigen-binding fragment thereof are used interchangeably herein and refer to the ability of the antibody or antigen-binding fragment to form one or more noncovalent bonds with a cognate antigen, by noncovalent interactions between the antibody combining site(s) of the antibody and the antigen.
  • the antigen can be an isolated antigen or presented in a virus.
  • an antibody that specifically binds to a virus antigen or virus is one that binds to the virus antigen (or to the antigen in the virus or to the virus) with an affinity constant (K a ) of about or IxIO 7 M “1 or Ix 10 8 M '1 or greater (or a dissociation constant (K ⁇ 0 of 1 x 10 '7 M or 1 x 10 "8 M or less).
  • affinity constants can be determined by standard kinetic methodology for antibody reactions, for example, immunoassays, surface plasmon resonance (SPR) (Rich and Myszka (2000) Curr. Opin. Biotechnol 11:54; Englebienne (1998) Analyst.
  • ITC isothermal titration calorimetry
  • other kinetic interaction assays known in the art (see, e.g., Paul, ed., Fundamental Immunology, 2nd ed., Raven Press, New York, pages 332-336 (1989); see also U.S. Pat. No. 7,229,619 for a description of exemplary SPR and ITC methods for calculating the binding affinity of anti-HCV antibodies).
  • Instrumentation and methods for real time detection and monitoring of binding rates are known and are commercially available (e.g., BiaCore 2000, Biacore AB, Upsala, Sweden and GE Healthcare Life Sciences; Malmqvist (2000) Biochem. Soc. Trans. 27:335).
  • an antibody that specifically binds to a virus antigen can bind to other peptides, polypeptides, or proteins or viruses with equal or lower binding affinity.
  • an antibody or antigen-binding fragment thereof provided herein that binds specifically binds to a HCV E2 protein does not cross-react with other antigens or cross reacts with substantially (at least 10-100 fold) lower affinity for such antigens.
  • an antibody or antigen-binding fragment thereof provided herein that binds specifically to a HCV E1E2 complex does not cross-react with other antigens or cross reacts with substantially (at least 10-100 fold) lower affinity for such antigens.
  • An antibody or antigen-binding fragment thereof provided herein that binds selectively to E2 typically also binds to the El E2 complex.
  • Antibodies or antigen- binding fragments that selectively bind to a particular virus antigen can be identified, for example, by immunoassays, such as radioimmunoassays (RIA), enzyme-linked immunosorbent assays (ELISAs), surface plasmon resonance, or other techniques known to those of skill in the art.
  • An antibody or antigen-binding fragment thereof that specifically binds to an epitope on a HCV E2 or El E2 typically is one that binds to the epitope (presented in the protein or virus) with a higher binding affinity than to any cross-reactive epitope as determined using experimental techniques, such as, but not limited to, immunoassays, surface plasmon resonance, or other techniques known to those of skill in the art.
  • a recombinantly produced protein such as HCV E1E2
  • the affinity for the antibody or antigen-binding fragments for virus or the antigen as presented in the virus can be determined.
  • the target such as the isolated protein or the virus, will be identified.
  • the term "conformation-dependent,” in reference to an antibody, means that the antibody recognizes and binds specifically with discontinuous epitopes composed of amino acid residues that are located at some distance from each other, i.e. the residues are discontinuous in the polypeptide sequence.
  • the discontinuous epitopes come together through proper folding of the polypeptide to form a binding site, i.e. a conformational epitope that is recognized by a conformation-dependent antibody.
  • cross-neutralizing means the ability to neutralize at least two HCV strains, isolates, species, quasispecies, subtypes or genotypes.
  • neutralize as used herein in reference to an antibody, means that the antibody can prevent or reduce HCV infection or replication in a cell culture or in a mammal, as well as alleviate one or more symptoms associated with HCV infection in a mammal.
  • reduce means a decrease in any amount such as a 5 %, 10 %, 15 %, 20 %, 25 %, 30 %, 35 %, 40 %, 45 %, 50 %, 55 %, 60 %, 65 % or more than 65 %.
  • HCV infection or replication can be detected by examining HCV RNA levels, virus particles count or clinical symptoms associated with HCV infection. Whether an antibody will prevent or reduce HCV infection or replication or alleviate associated symptoms can be determined using methods known in the art, as well as the methods described herein, including determining the level of HCV RNA in a sample from a mammal that has been infected with HCV or detecting reduction of signals from a reporter gene encoded by the virus such as, for example, the relative light unit (RLU) for luciferase or the mean fluorescence intensity (MFI) of green fluorescent protein (GFP).
  • RLU relative light unit
  • MFI mean fluorescence intensity
  • GFP green fluorescent protein
  • linker or “spacer” peptide refers to short sequences of amino acids, such as 2, 5 or 10 to 20, 30, 40, 50, 60, 70 or 80, that join two polypeptide sequences (or nucleic acid encoding such an amino acid sequence).
  • Peptide linker refers to the short sequence of amino acids joining the two polypeptide sequences. Linkers are well-known and any known linkers can be used in the provided methods. Exemplary of polypeptide linkers are (Gly-Ser) n amino acid sequences, with some GIu or Lys residues dispersed throughout to increase solubility. Other exemplary linkers are described herein; any of these and other known linkers can be used with the provided compositions and methods.
  • Linkers with reference to the mutant E2 polypeptides also include all or part of the native sequence joining the segments in the native E2 polypeptide, as long as the resulting polypeptide does not correspond to amino acids about 412-462 of the E2 polypeptide of the particular HCV isolate.
  • a "tag” or an “epitope tag” refers to a sequence of amino acids, typically added to the N- or C- terminus of a polypeptide, such as the polypeptides provided herein.
  • tags fused to a polypeptide can facilitate polypeptide purification and/or detection.
  • a tag or tag polypeptide refers to a polypeptide that has enough residues to provide an epitope recognized by an antibody or can serve for detection or purification, yet is short enough such that it does not interfere with activity of the chimeric polypeptide to which it is linked.
  • the tag polypeptide typically is sufficiently unique so an antibody that specifically binds thereto does not substantially cross-react with epitopes in the polypeptide to which it is linked. Suitable tag polypeptides generally have at least 5 or 6 amino acid residues and usually between about 8-50 amino acid residues, typically between 9-30 residues.
  • the tags can be linked to one or more chimeric polypeptides in a multimer and permit detection of the multimer or its recovery from a sample or mixture. Such tags are well known and can be readily synthesized and designed.
  • Exemplary tag polypeptides include those used for affinity purification and include, histidine (His) tags, the influenza hemagglutinin (HA) tag polypeptide and its antibody 12CA5, (Field et al.
  • nucleic acid refers to a polymer of deoxyribose nucleic acids (DNA), as well as ribose nucleic acids (RNA).
  • the term includes linear molecules, as well as covalently closed circular molecules. It includes single stranded molecules, as well as double stranded molecules.
  • Nucleic acids also include DNA and RNA derivatives containing, for example, a nucleotide analog or a "backbone" bond other than a phosphodiester bond, for example, a phosphotriester bond, a phosphoramidate bond, a phosphorothioate bond, a thioester bond, or a peptide bond (peptide nucleic acid).
  • RNA or DNA made from nucleotide analogs, single (sense or antisense) and double-stranded nucleic acids.
  • Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine and deoxythymidine.
  • RNA the uracil base is undine.
  • Nucleic acids can contain nucleotide analogs, including, for example, mass modified nucleotides, which allow for mass differentiation of nucleic acid molecules; nucleotides containing a detectable label such as a fluorescent, radioactive, luminescent or chemiluminescent label, which allow for detection of a nucleic acid molecule; or nucleotides containing a reactive group such as biotin or a thiol group, which facilitates immobilization of a nucleic acid molecule to a solid support.
  • a nucleic acid also can contain one or more backbone bonds that are selectively cleavable, for example, chemically, enzymatically or photolytically cleavable.
  • a nucleic acid can include one or more deoxyribonucleotides, followed by one or more ribonucleotides, which can be followed by one or more deoxyribonucleotides, such a sequence being cleavable at the ribonucleotide sequence by base hydrolysis.
  • a nucleic acid also can contain one or more bonds that are relatively resistant to cleavage, for example, a chimeric oligonucleotide primer, which can include nucleotides linked by peptide nucleic acid bonds and at least one nucleotide at the 3' end, which is linked by a phosphodiester bond or other suitable bond, and is capable of being extended by a polymerase.
  • Peptide nucleic acid sequences can be prepared using well-known methods (see, for example, Weiler et al. (1997) Nucleic Acids Res. 25:2792-2799).
  • polynucleotide and “nucleic acid molecule” refer to an oligomer or polymer containing at least two linked nucleotides or nucleotide derivatives, including a deoxyribonucleic acid (DNA) and a ribonucleic acid (RNA), joined together, typically by phosphodiester linkages.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • Polynucleotides also include DNA and RNA derivatives containing, for example, a nucleotide analog or a "backbone" bond other than a phosphodiester bond, for example, a phosphotriester bond, a phosphoramidate bond, a phosphorothioate bond, a thioester bond, or a peptide bond (peptide nucleic acid).
  • Polynucleotides include single-stranded and/or double-stranded polynucleotides, such as deoxyribonucleic acid (DNA), and ribonucleic acid (RNA) as well as analogs or derivatives of either RNA or DNA.
  • RNA or DNA made from nucleotide analogs, single (sense or antisense) and double-stranded polynucleotides.
  • Deoxyribonucleotides include deoxyadenosine, deoxycytidine, deoxyguanosine and deoxythymidine.
  • RNA the uracil base is undine.
  • Polynucleotides can contain nucleotide analogs, including, for example, mass modified nucleotides, which allow for mass differentiation of polynucleotides; nucleotides containing a detectable label such as a fluorescent, radioactive, luminescent or chemiluminescent label, which allow for detection of a polynucleotide; or nucleotides containing a reactive group such as biotin or a thiol group, which facilitates immobilization of a polynucleotide to a solid support.
  • a polynucleotide also can contain one or more backbone bonds that are selectively cleavable, for example, chemically, enzymatically or photolytically cleavable.
  • a polynucleotide can include one or more deoxyribonucleotides, followed by one or more ribonucleotides, which can be followed by one or more deoxyribonucleotides, such a sequence being cleavable at the ribonucleotide sequence by base hydrolysis.
  • a polynucleotide also can contain one or more bonds that are relatively resistant to cleavage, for example, a chimeric oligonucleotide primer, which can include nucleotides linked by peptide nucleic acid bonds and at least one nucleotide at the 3' end, which is linked by a phosphodiester bond or other suitable bond, and is capable of being extended by a polymerase.
  • Peptide nucleic acid sequences can be prepared using well-known methods (see, for example, Weiler et al. (1997) Nucleic Acids Res. 25:2792-2799).
  • Exemplary of the nucleic acid molecules (polynucleotides) provided herein are oligonucleotides, including synthetic oligonucleotides, oligonucleotide duplexes, primers, including fill- in primers, and oligonucleotide duplex cassettes.
  • DNA construct is a single or double stranded, linear or circular DNA molecule that contains segments of DNA combined and juxtaposed in a manner not found in nature. DNA constructs exist as a result of human manipulation, and include clones and other copies of manipulated molecules.
  • a "DNA segment” is a portion of a larger DNA molecule having specified attributes.
  • a DNA segment encoding a specified polypeptide is a portion of a longer DNA molecule, such as a plasmid or plasmid fragment, which, when read from the 5' to 3' direction, encodes the sequence of amino acids of the specified polypeptide.
  • a positive strand polynucleotide refers to the "sense strand" of a polynucleotide duplex, which is complementary to the negative strand or the "antisense” strand.
  • the sense strand is the strand that is identical to the mRNA strand that is translated into a polypeptide, while the antisense strand is complementary to that strand.
  • Positive and negative strands of a duplex are complementary to one another.
  • isolated as used herein with reference to a nucleic acid molecule, means that the nucleic acid molecule is free of unrelated nucleic acid sequences, i.e.
  • nucleic acid sequences encoding other genes or non-E2 polypeptide sequences, or those involved in the expression of such other genes, that flank it's 5' and 3' ends in the naturally-occurring genome of the organism from which the nucleic acid provided herein is derived. Accordingly, an "isolated nucleic acid” provided herein has a structure that is different from that of any naturally occurring nucleic acid or to that of any fragment of a naturally occurring genomic nucleic acid spanning more than three separate genes.
  • isolated nucleic acid molecule includes, for example, (1) a DNA molecule that has the sequence of part of a naturally occurring genomic DNA molecule, but is not flanked by both of the coding sequences that flank that part of the molecule in the genome of the organism in which it naturally occurs; (2) a nucleic acid incorporated into a vector or into the genomic DNA of a prokaryote or eukaryote in a manner such that the resulting molecule is not identical to any naturally-occurring vector or genomic DNA; (3) a separate molecule such as a cDNA, a genomic fragment, a fragment produced by polymerase chain reaction (PCR), or a restriction fragment; and (4) a recombinant nucleotide sequence that is part of a hybrid gene, i.e.
  • PCR polymerase chain reaction
  • nucleic acids present in mixtures of (1) DNA molecules, (2) transfected cells, and (3) cell clones, e.g., as these occur in a DNA library such as a cDNA or genomic DNA library.
  • a regulatory region or an "expression control sequence" of a nucleic acid molecule means a cis-acting nucleotide sequence that influences expression, positively or negatively, of an operatively linked gene.
  • Regulatory regions include sequences of nucleotides that confer inducible (i.e., require a substance or stimulus for increased transcription) expression of a gene. When an inducer is present or at increased concentration, gene expression can be increased. Regulatory regions also include sequences that confer repression of gene expression (i.e., a substance or stimulus decreases transcription). When a repressor is present or at increased concentration gene expression can be decreased.
  • Regulatory regions are known to influence, modulate or control many in vivo biological activities including cell proliferation, cell growth and death, cell differentiation and immune modulation. Regulatory regions typically bind to one or more trans-acting proteins, which results in either increased or decreased transcription of the gene.
  • Promoters are sequences located around the transcription or translation start site, typically positioned 5' of the translation start site. Promoters usually are located within 1 Kb of the translation start site, but can be located further away, for example, 2 Kb, 3 Kb, 4 Kb, 5 Kb or more, up to and including 10 Kb. Enhancers are known to influence gene expression when positioned 5' or 3' of the gene, or when positioned in or as part of an exon or an intron. Enhancers also can function at a significant distance from the gene, for example, at a distance from about 3 Kb, 5 Kb, 7 Kb, 10 Kb, 15 Kb or more.
  • Regulatory regions or expression control sequences also include, in addition to promoter regions, sequences that facilitate translation, splicing signals for introns, maintenance of the correct reading frame of the gene to permit in-frame translation of mRNA and stop codons, leader sequences and fusion partner sequences, internal ribosorne binding site (IRES) elements for the creation of multigene, or polycistronic, messages, polyadenylation signals to provide proper polyadenylation of the transcript of a gene of interest and stop codons, and can be optionally included in an expression vector.
  • IRIS internal ribosorne binding site
  • a "host cell” is a cell that is used to receive, maintain, reproduce and amplify a vector.
  • a host cell also can be used to express the polypeptide encoded by the vector.
  • the nucleic acid contained in the vector is replicated when the host cell divides, thereby amplifying the nucleic acids.
  • the host cell is a genetic package, which can be induced to express the variant polypeptide on its surface.
  • the host cell is infected with the genetic package.
  • the host cells can be phage-display compatible host cells, which can be transformed with phage or phagemid vectors and accommodate the packaging of phage expressing fusion proteins containing the variant polypeptides.
  • a "vector" is a replicable nucleic acid from which one or more heterologous proteins can be expressed when the vector is transformed into an appropriate host cell.
  • Reference to a vector includes those vectors into which a nucleic acid encoding a polypeptide or fragment thereof can be introduced, typically by restriction digest and ligation.
  • Reference to a vector also includes those vectors that contain nucleic acid encoding a polypeptide.
  • the vector is used to introduce the nucleic acid encoding the polypeptide into the host cell for amplification of the nucleic acid or for expression/display of the polypeptide encoded by the nucleic acid.
  • the vectors typically remain episomal, but can be designed to effect integration of a gene or portion thereof into a chromosome of the genome.
  • vectors that are artificial chromosomes such as yeast artificial chromosomes and mammalian artificial chromosomes. Selection and use of such vehicles are well known to those of skill in the art.
  • a vector also includes "virus vectors” or “viral vectors.”
  • viral vectors are engineered viruses that are operatively linked to exogenous genes to transfer (as vehicles or shuttles) the exogenous genes into cells.
  • an "expression vector” is a nucleic acid molecule capable of transporting and/or allowing for the expression of another nucleic acid to which it has been linked.
  • Expression vectors contain appropriate expression control sequences that direct expression of a nucleic acid that is operably linked to the expression control sequence to produce a transcript.
  • the product of that expression is referred to as a messenger ribose nucleic acid (mRNA) transcript.
  • mRNA messenger ribose nucleic acid
  • the expression vector also can include other sequences such as enhancer sequences, synthetic introns, adenovirus tripartite leader (TPL) sequences and modified polyadenylation and transcriptional termination sequences, e.g. bovine growth hormone or rabbit beta-globulin polyadenylation sequences, to improve expression of the nucleic acid encoding the E2 polypeptide.
  • TPL adenovirus tripartite leader
  • expression refers to the process by which polypeptides are produced by transcription and translation of polynucleotides.
  • the level of expression of a polypeptide can be assessed using any method known in art, including, for example, methods of determining the amount of the polypeptide produced from the host cell. Such methods can include, but are not limited to, quantitation of the polypeptide in the cell lysate by ELISA 5 Coomassie blue staining following gel electrophoresis, Lowry protein assay and Bradford protein assay.
  • the term "purified" with reference to a polypeptide or antibody preparation means that the polypeptide or antibody in the preparation is substantially free of naturally-associated components, i.e. components that accompany it in its natural state.
  • the term “purified” also encompasses a biological sample such as a blood sample that has been subject to at least one separation step, for example, centrifugation to separate cellular components from non- cellular components.
  • a polypeptide provided herein can constitute at least about 25 % by weight of a sample containing the polypeptide provided herein, and usually constitutes at least about 50%, at least about 75 %, at least about 85 %, at least about 90 % of a sample, particularly at least about 95 % of the sample or 99 % or more.
  • prevent refers to use in a prophylactic manner that includes, for example, preventing a new infection or viral replication or reducing the probability of infection, as well as preventing the onset of symptoms and/or their underlying cause.
  • the terms “treat,” “treating” and “treatment,” include reducing viral replication, reducing the severity and/or frequency of symptoms, eliminating the symptoms and/or underlying cause or improving or remediating damage associated with the infection.
  • reduce means a decrease in any amount, for example, a decrease of 5 %, 10 %, 20 %, 40 %, 50 %, 60 %, 70 %, 80 %, 90 % or more than 90 %.
  • a "pharmaceutically effective agent” includes any therapeutic agent or bioactive agents, including, but not limited to, for example, anesthetics, vasoconstrictors, dispersing agents, conventional therapeutic drugs, including small molecule drugs and therapeutic proteins.
  • a "therapeutic effect” means an effect resulting from treatment of a subject that alters, typically improves or ameliorates the symptoms of a disease or condition or that cures a disease or condition.
  • a “therapeutically effective amount” or a “therapeutically effective dose” refers to the quantity of an agent, compound, material, or composition containing a compound that is at least sufficient to produce a therapeutic effect following administration to a subject. Hence, it is the quantity necessary for preventing, curing, ameliorating, arresting or partially arresting a symptom of a disease or disorder.
  • therapeutic efficacy refers to the ability of an agent, compound, material, or composition containing a compound to produce a therapeutic effect in a subject to whom the agent, compound, material, or composition containing a compound has been administered.
  • a prophylactically effective amount or a “prophylactically effective dose” refers to the quantity of an agent, compound, material, or composition containing a compound that when administered to a subject, will have the intended prophylactic effect, e.g., preventing or delaying the onset, or reoccurrence, of disease or symptoms, reducing the likelihood of the onset, or reoccurrence, of disease or symptoms, or reducing the incidence of viral infection.
  • the full prophylactic effect does not necessarily occur by administration of one dose, and can occur only after administration of a series of doses.
  • a prophylactically effective amount can be administered in one or more administrations.
  • amelioration of the symptoms of a particular disease or disorder by a treatment refers to any lessening, whether permanent or temporary, lasting or transient, of the symptoms that can be attributed to or associated with administration of the composition or therapeutic.
  • diagnosisically effective amount refers to the quantity of an agent, compound, material, or composition containing a detectable compound that is at least sufficient for detection of the compound following administration to a subject.
  • a diagnostically effective amount of an anti- RSV antibody or antigen-binding fragment thereof such as a detectably-labeled antibody or antigen-binding fragment thereof or an antibody or antigen-binding fragment thereof that can be detected by a secondary agent, administered to a subject for detection is the quantity of the antibody or antigen-binding fragment thereof which is sufficient to enable detection of the site having the HCV antigen for which the antibody or antigen-binding fragment thereof is specific.
  • a detectably labeled antibody or antigen-binding fragment thereof is given in a dose which is diagnostically effective.
  • a label or detectable moiety is a detectable marker (e.g., a fluorescent molecule, chemiluminescent molecule, a bioluminescent molecule, a contrast agent (e.g., a metal), a radionuclide, a chromophore, a detectable peptide, or an enzyme that catalyzes the formation of a detectable product) that can be attached or linked directly or indirectly to a molecule or associated therewith and can be detected in vivo and/or in vitro.
  • a detectable marker e.g., a fluorescent molecule, chemiluminescent molecule, a bioluminescent molecule, a contrast agent (e.g., a metal), a radionuclide, a chromophore, a detectable peptide, or an enzyme that catalyzes the formation of a detectable product
  • a detectable marker e.g., a fluorescent molecule, chemiluminescent molecule
  • the detection method can be any method known in the art, including known in vivo and/or in vitro methods of detection (e.g., imaging by visual inspection, magnetic resonance (MR) spectroscopy, ultrasound signal, X-ray, gamma ray spectroscopy (e.g., positron emission tomography (PET) scanning, single-photon emission computed tomography (SPECT)), fluorescence spectroscopy or absorption).
  • Indirect detection refers to measurement of a physical phenomenon, such as energy or particle emission or absorption, of an atom, molecule or composition that binds directly or indirectly to the detectable moiety.
  • the term "subject" refers to an animal, including a mammal, such as a human being.
  • a patient refers to a human subject.
  • animal includes any animal, such as, but are not limited to primates including humans, gorillas and monkeys; rodents, such as mice and rats; fowl, such as chickens; ruminants, such as goats, cows, deer, sheep; ovine, such as pigs and other animals.
  • rodents such as mice and rats
  • fowl such as chickens
  • ruminants such as goats, cows, deer, sheep
  • ovine such as pigs and other animals.
  • Non-human animals exclude humans as the contemplated animal.
  • the enzymes provided herein are from any source, animal, plant, prokaryotic and fungal. Most enzymes are of animal origin, including mammalian origin.
  • pharmaceutically acceptable it is meant a carrier, diluent, excipient, and/or salt that is compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof, for example, a buffered aqueous, oil or organic medium containing optional stabilizing agents and adjuvants for stimulation of immune binding.
  • unit dose form refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art.
  • a single dosage formulation refers to a formulation for direct administration.
  • an “article of manufacture” is a product that is made and sold.
  • Fluids refers to any composition that can flow. Fluids thus encompass compositions that are in the form of semi-solids, pastes, solutions, aqueous mixtures, gels, lotions, creams and other such compositions.
  • composition refers to any mixture. It can be a solution, suspension, liquid, powder, paste, aqueous, non-aqueous or any combination thereof.
  • a "combination" refers to any association between or among two or more items.
  • the combination can be two or more separate items, such as two compositions or two collections, can be a mixture thereof, such as a single mixture of the two or more items, or any variation thereof.
  • the elements of a combination are generally functionally associated or related.
  • combination therapy refers to administration of two or more different therapeutics, such as two or more different anti-RSV antibodies and/or anti- RSV antibodies and antigen-binding fragments thereof.
  • the different therapeutic agents can be provided and administered separately, sequentially, intermittently, or can be provided in a single composition.
  • kits are packaged combinations that optionally includes other elements, such as additional reagents and instructions for use of the combination or elements thereof, for a purpose including, but not limited to, activation, administration, diagnosis, and assessment of a biological activity or property.
  • the singular forms "a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.
  • reference to a polypeptide, comprising "an immunoglobulin domain” includes polypeptides with one or a plurality of immunoglobulin domains.
  • the term “or” is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive.
  • ranges and amounts can be expressed as “about” a particular value or range. About also includes the exact amount. Hence “about 5 amino acids” means “about 5 amino acids” and also “5 amino acids.”
  • “optional” or “optionally” means that the subsequently described event or circumstance does or does not occur and that the description includes instances where said event or circumstance occurs and instances where it does not.
  • an optionally variant portion means that the portion is variant or non- variant.
  • the abbreviations for any protective groups, amino acids and other compounds are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (see, Biochem.
  • mutant E2 polypeptides Provided herein are mutant E2 polypeptides, compositions containing them, as well as methods for their production, purification and use.
  • the polypeptides provided herein can be used as immunogens to elicit antibodies that can protect against infection by a hepatitis C virus (HCV).
  • HCV E2 polypeptides provided herein display the conserved neutralizing AR3 epitopes recognized by conformation- dependent cross-neutralizing anti-HCV antibodies.
  • mutant E2 polypeptides displaying conserved neutralizing epitopes, nucleic acids encoding these polypeptides and expression vectors for their production.
  • cells comprising such nucleic acids or expression vectors, a preparation or pharmaceutical composition comprising a mutant HCV E2 polypeptide, as well as (1) a method of eliciting an immune response in a mammal comprising administering a mutant HCV E2 polypeptide, (2) a method for determining whether a mammal has been infected with an HCV, and (3) a method for identifying an anti-HCV agent.
  • a preparation or pharmaceutical composition comprising a mutant HCV E2 polypeptide
  • Hepatitis C virus is a noncytopathic, positive-stranded RNA virus belonging to the Hepacivirus genus of the Flaviviridae family that causes acute and chronic hepatitis and hepatocellular carcinoma (Hoofhagle, J. H. (2002) Hepatology 36, S21-29).
  • the hepatocyte is the primary target cell, although various lymphoid populations, especially B cells and dendritic cells also can be infected at lower levels (Kanto et al, Immunol. 162, 5584-5591 (1999); Auffermann-Gretzinger et al, Blood 97, 3171-3176 (2001); Hiasa et al. (1998) Biochem. Biophys. Res. Commun.
  • HCV infection A striking feature of HCV infection is its tendency towards chronicity with at least 70 % of acute infections progressing to persistence (Hoofhagle, J. H. (2002) Hepatology 36, S21-29). HCV chronicity is often associated with significant liver disease, including chronic active hepatitis, cirrhosis and hepatocellular carcinoma (Alter, H. J. & Seeff, L. B. (2000) Semin. Liver Dis. 20, 17-35). With over 170 million people currently infected (id.), HCV represents a growing public health concern.
  • HCV viruses can be categorized into several genotypes and subtypes.
  • Exemplary HCV genotypes include, but are not limited to, genotype 1, 2, 3, 4, 5 and 6.
  • Exemplary of HCV subtypes include, but are not limited to, Ia, Ib, Ic, 2a, 2b, 2c, 2i, 2k, 3a, 3b, 3k, 4a, 4d, 4f, 5a, 6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 61, 6m, 6n, 6o, 6q, 6p and 6t .
  • the single stranded HCV RNA genome has a single open reading frame (ORF) encoding a large polyprotein.
  • the polyprotein has about 3010-3033 amino acids (Q.-L. Choo, etal. Proc. Natl. Acad. ScL USA 88, 2451-2455 (1991); N. Kato et al., Proc. Natl. Acad. Sci. USA 87, 9524-9528 (1990); A. Takamizawa et al., J. Virol. 65, 1105-1113 (1991)).
  • Nucleic acid and amino acid sequences for different isolates of HCV can be found in the art, for example, in the National Center for Biotechnology Information (NCBI) database (see ncbi.nlm.nih.gov).
  • HCV subtype 1 a is strain H77, which can be found in the NCBI database as accession number AF009606. Its polyprotein sequence (AAB66324) is as follows:
  • HCV subtype Ib is strain HCV-L2, which can be found in the NCBI database as accession number UOl 214 (gi 437107). Its polyprotein sequence (AAA75355 ) is as follows:
  • HCV polyprotein sequences are known in the art and can be used to generate the mutant HCV E2 polypeptide provided herein (see for example, hcvdb.org/viruses.asp; .ncbi.nlm.nih.gov; and hcv.lanl.gov and HCV sequence databases referenced in Kuiken et al. (2005) Bioinformatics 21 (3):379-84 and Yusim et al. (2005) Applied Bioinformatics 4(4)). Additional examples include a Taiwan isolate of hepatitis C virus available in the NCBI database at accession number P29846 (gi: 266821).
  • HCV polyprotein sequences include, but are not limited to, NCBI accession number AF009606, AY734971, AJ238799, AY545953, AY734974, AB047639, AFl 77036, AY734977, AY734982, AY734984, AY734987, EF427672, and AY736194.
  • NCBI accession number AF009606, AY734971, AJ238799, AY545953, AY734974, AB047639, AFl 77036, AY734977, AY734982, AY734984, AY734987, EF427672, and AY736194 C. Mutant E2 Polypeptides
  • mutant HCV E2 polypeptides differs from a naturally-occurring E2 polypeptide of HCV in that the mutant E2 polypeptide provided herein has amino acid deletions relative to the naturally-occurring E2 polypeptide.
  • Exemplary regions of an E2 polypeptide that can be removed include, for example, the region defined by amino acid residues corresponding to 384-411, 460 to 485, 570-580, 646-647, 648-661, 662-717 or 718- 746, (the amino acid positions corresponding to the amino acid positions in an HCV polyprotein) or any combination thereof can be deleted from a naturally-occurring E2 polypeptide of HCV to generate a mutant E2 polypeptide provided herein.
  • a mutant E2 polypeptide provided herein thus has an amino acid sequence that comprises, from the amino to the carboxy termini: (1) a first segment that corresponds to amino acid residues 412 to 459 of a select hepatitis C virus polyprotein, (2) a second segment that corresponds to amino acid residues 486 to 569 of the select hepatitis C virus polyprotein, and (3) a third segment that corresponds to amino acid residues 581 to 645 of the select hepatitis C virus polyprotein.
  • one or more immunodominant epitopes in the naturally-occurring E2 polypeptide are eliminated or its immunogenicity to particular epitopes attenuated, while the immunogenicity of conserved or cross-neutralizing epitopes are augmented.
  • immunodominant epitopes such as, for example, the hypervariable region 1 (amino acid residues 384 to 412) or the epitopes recognized by the ARIA and ARlB antibodies that include the residues T416, T416, N417, R483, P484, Y485, V538, N540, P544, P545, G547 and W549.
  • mutant E2 polypeptide provided herein also can have one or more amino acid substitutions or deletions at positions 416, 417, 483, 484, 485, 538, 540, 544, 545, 547, 549 or any combinations thereof relative to the E2 polypeptide sequence of HCV (e.g. corresponding to the HCV strain H77).
  • the segments of the mutant E2 polypeptide can be linked directly or indirectly, in any order, via a linker provided that the polypeptide properly folds to present the conserved HCV E2 conformational epitope.
  • linkers are known in the art and include, for example, polypeptide linkers.
  • the polypeptide linker can link the first and second segments, and the polypeptide linker is at least 10 amino acids, e.g.
  • the polypeptide linker can link the second and third segments, and the polypeptide linker is about 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or more than 28 amino acid residues in length provided that the polypeptide properly folds to form the conserved HCV E2 conformational epitope.
  • the first and second segments are linked via a polypeptide linker which is about 26 amino acid residues
  • the second and third segments are linked via a polypeptide linker which is about 12 amino acid residues in length.
  • any HCV E2 polypeptide can be modified to produce the mutant E2 polypeptides provided herein.
  • the amino acid segments can be derived for an HCV E2 polypeptide of any genotype (e.g. 1, 2, 3, 4, 5, or 6) or subtype (e.g. Ia, Ib, Ic, 2a, 2b, 2c, 2i, 2k, 3a, 3b, 3k, 4a, 4d, 4f, 5a, 6a, 6b, 6c, 6d, 6e, 6f, 6g, 6h, 6i, 6j, 6k, 61, 6m, 6n, 6o, 6q, 6p or 6t).
  • Exemplary naturally-occurring HCV E2 sequences that can be modified include, but are not limited to, HCV E2 polypeptides shown in Table 2 below.
  • amino acid sequence segments from select hepatitis C viruses that can be employed to produce the mutant E2 polypeptides provided herein include, but are not limited, to the segments that correspond to the polypeptide segments shown in Table 3.
  • the first segment of the mutant E2 polypeptide provide herein is 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % identical to amino . acids 412 to 459 of a hepatitis C virus E2 polypeptide, such as H77.
  • the second segment of the mutant E2 polypeptide is 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % identical to amino acids 486 to 569 of a hepatitis C virus E2 polypeptide, such as H77.
  • the third segment of the mutant E2 polypeptide provided herein is 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % identical to amino acids 581 to 645 of a hepatitis C virus E2 polypeptide, such as H77.
  • the first amino acid segment of the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 888-912 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity to any one of SEQ ID NOS: 888-912.
  • the second amino acid segment of the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 913-937 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity to any one of SEQ ID NOS: 913- 937.
  • the third amino acid segment of the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 938-962 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %, 97 %, 98 %, or 99 % sequence identity to any one of SEQ ID NOS: 938-962.
  • the mutant E2 polypeptide has the sequence of any one of SEQ ID NOS: 727 '-730 and 740-742 or has 65 %, 70 %, 75 %, 80 %, 85 %, 90 %, 91 %, 92 %, 93 %, 94 %, 95 %, 96 %,
  • Exemplary mutant E2 polypeptides provided herein differ from the corresponding naturally-occurring E2 amino acid sequence in that the mutant E2 polypeptide provided herein does not include one or more segments defined by (1 ) amino acid residues 384 to 411 of the hypervariable region 1 of the naturally- occurring E2 polypeptide; (2) amino acid residues 460 to 485; (3) amino acid residues 570-580, (4) amino acid residues 647-661, (5) amino acid residues 662-717 or (6) 718-746, or any combination thereof.
  • the mutant E2 polypeptide provided herein also can differ from the corresponding naturally-occurring E2 amino acid sequence in that the mutant E2 polypeptide can have at least one amino acid substitution at position 416, 417, 483, 484, 485, 538, 540, 544, 545, 547, 549 or any combinations thereof.
  • a mutant E2 polypeptide provided herein can have at least two amino acid substitutions at these positions, e.g. 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acid substitutions.
  • the amino acid that can be substituted at these positions can be one that has a different chemical or physical property from the naturally-occurring residue.
  • the proline residues at position 484, 544 or 545 can be substituted with an amino acid residue that enable the polypeptide to be more flexible such as for example an alanine, valine or other non-cyclic residues.
  • the glycine residue at position 547 can be substituted with an amino acid residue that has a bulkier side chain such as, for example, valine, leucine, methionine, phenylalanine, tyrosine, tryptophan, histidine, lysine, arginine, aspartic acid, glutamic acid, asparagine or glutamine, while the tryptophan residue at position 549 can be substituted with an amino acid residue that has a less bulky side chain, for example, glycine, alanine, valine, serine, cysteine, or threonine.
  • the threonine residue at position 416 can be substituted with a residue that does not have a hydroxyl or sulfur-containing side chain.
  • the acidic asparagine residue at position 417 or 540 can be substituted with, for example, a basic amino acid residue such as histidine, lysine or arginine, while the basic arginine residue at position 483, for example, can be substituted with, for example, an acidic residue such as aspartic acid, glutamic acid, asparagine or glutamine.
  • the aromatic amino acid tyrosine at position 485 can be substituted with, for example, a non-aromatic residue, while the valine at position 538 can be substituted with a residue having a bulkier side chain, a basic or acidic residue, or one with an aromatic, hydroxyl or sulfur-containing side chain.
  • An exemplary substitution or combination of substitutions is one that decreases the immunogenicity or function of epitopes recognized by the ARl antibodies such as ARIA and ARlB.
  • the mutant E2 polypeptide provided herein also can have one or more other substitutions, insertions or deletions relative to a naturally-occurring E2 polypeptide as long as the mutant E2 polypeptide sequence includes the discontinuous epitopes described herein that come together to form a conformational epitope recognized by a conformation-dependent cross-neutralizing antibody such as the AR3A, AR3B, AR3C or AR3D antibody.
  • the mutant E2 polypeptide provided herein contain one or more discontinuous epitopes of an E2 polypeptide.
  • the polypeptides provided herein include the following amino acid regions: (1) amino acid residues 412 to 424; (2) amino acid residues 436 to 447; and (3) amino acid residues 523 to 540 relative to the naturally- occurring E2 polypeptide (e.g. HCV strain H77 polypeptide and other HCV strains, isolates, species, quasispecies, subtypes or genotypes). Sequences of the discontinuous epitopes can be determined based on sequence alignment of the HCV E2 or HCV polyprotein sequence with the sequence of strain H77 using the methods described above.
  • mutant E2 polypeptides provided herein include the mutant E2 polypeptides shown in Table 5 below.
  • a polypeptide provided herein also can include non-E2 sequences at the N or C terminus.
  • Non-E2 sequences can be, for example, a tag such as an N-terminal ubiquitin signal, a poly-histidine sequence (SEQ ID NO:685), a FLAG sequence (SEQ ID NO:607), an HA sequence (SEQ ID NO:608), a myc sequence (SEQ ID NO:609), a V5 sequence (SEQ ID NO:610), a chitin binding protein sequence, a maltose binding protein sequence (SEQ ID NO:687) or a glutathione-S-transferase sequence.
  • D. Nucleic Acids Encoding Mutant E2 Polypeptides Provided herein are isolated nucleic acids encoding modified E2 polypeptides.
  • Nucleic acids encoding mutant E2 polypeptides provided herein can be generated from nucleic acids encoding the naturally-occurring HCV polyprotein using methods known to those skilled in the art. For example, nucleic acids encoding mutant E2 polypeptides containing various amino acid substitutions can be produced by site- specific mutagenesis and polymerase chain reaction (PCR) amplification from the nucleic acids encoding the naturally-occurring HCV polyprotein. Nucleic acids encoding mutant E2 polypeptides, i.e.
  • polypeptides that do not include amino acid residues 384 to 410 of the hypervariable region of the naturally occurring E2 protein can be produced by PCR using primers that do not encompass the nucleotides coding for amino acid residues 384 to 410.
  • Nucleic acid sequences encoding the naturally- occurring HCV polyproteins are disclosed at the NCBI website (ncbi.nlm.nih.gov).
  • Selected accession numbers for nucleic acids encoding the naturally-occurring HCV polyproteins are as follows: AF009606; D10749; U01214; AY051292; AY746460; AY232731; D50409; DQ155561; AB031663; DQ437509; D49374; D63821; Yl 1604; DQ516083; EF589160; AF064490; AY859526; NC009827; EF420130; DQ314805 ; DQ835764; D63822; D84264; DQ835763; and DQ278894.
  • Nucleic acids encoding a polypeptide provided herein can be used for recombinant expression of the E2 polypeptide provided herein. Nucleic acids encoding a polypeptide provided herein also can be used in a nucleic acid-based vaccine to elicit an immune response against an HCV.
  • Nucleic acid encoding a polypeptide provided herein can be operably-linked to an expression control sequence in an expression vector, which can be introduced into a host cell for expression of the encoded polypeptide or administered to a mammal to elicit an immune response against the polypeptide.
  • nucleic acid sequences encoding mutant E2 polypeptides provided herein are shown in Table 6 below.
  • Nucleic acids encoding E2 polypeptides provided herein can be incorporated into viral, bacterial, insect, yeast or mammalian expression vectors.
  • nucleic acids encoding E2 polypeptides can be operably-linked to expression control sequences such as viral, bacterial, insect, yeast or mammalian promoters and enhancers.
  • expression control sequences such as enhancers and promoters include viral promoters such as SV 40 promoter, Rous Sarcoma Virus (RSV) promoter, and cytomegalovirus (CMV) immediate early promoter.
  • viral vectors include retrovirus-based vectors, e.g. lentiviruses, adenoviruses and adeno-associated viruses.
  • nucleic acid encoding an E2 polypeptide provided herein also can be linked to nucleic acid sequences that code for unrelated amino acid sequences such as N-terminal ubiquitin signals to improve antigen targeting, a poly-histidine sequence, a FLAG (DYKDDDDK, SEQ ID NO:607) sequence, an HA sequence, a myc sequence, a V5 sequence, a chitin binding protein sequence, a maltose binding protein sequence or a glutathione-S-transferase sequence.
  • unrelated amino acid sequences such as N-terminal ubiquitin signals to improve antigen targeting, a poly-histidine sequence, a FLAG (DYKDDDDK, SEQ ID NO:607) sequence, an HA sequence, a myc sequence, a V5 sequence, a chitin binding protein sequence, a maltose binding protein sequence or a glutathione-S-transferase sequence.
  • Expression vectors containing nucleic acids encoding E2 polypeptides can be introduced into bacterial, insect, yeast or mammalian host cells (e.g. CHO, Balb/3T3, HeLa 3 MT2, mouse NSO (non-secreting) and other myeloma cell lines, hybridoma and heterohybridoma cell lines, lymphocytes, fibroblasts, Sp2/0, COS, NIH3T3, HEK293 (e.g. 293T), 293S, 2B8, and HKB cells) for expression using conventional methods including, without limitation, transformation, transduction and transfection.
  • mammalian host cells e.g. CHO, Balb/3T3, HeLa 3 MT2, mouse NSO (non-secreting) and other myeloma cell lines, hybridoma and heterohybridoma cell lines, lymphocytes, fibroblasts, Sp2/0, COS, NIH3T3, HEK293 (e.g. 293T), 2
  • Expression vectors containing nucleic acids encoding E2 polypeptides, in saline for example can be introduced into a mammal, e.g. mammalian tissues, using standard methods including, for example, injection using a standard hypodermic needle, by a gene gun delivery, jet injection or liposome-mediated delivery. Injection can be intramuscular or intradermal. Electroporation, myotoxins such as bupivacaine or hypertonic solutions of saline or sucrose also can aid in delivery.
  • E2 polypeptides provided herein When expressed in bacterial, yeast, insect or mammalian host cells, E2 polypeptides provided herein can be purified using a method provided herein. Specifically, E2 polypeptides provided herein are purified by affinity chromatography using a cross-neutralizing antibody such as, for example, AR3A, AR3B, AR3C or AR3D in combination with size exclusion chromatography. More specifically, an E2 polypeptide provided herein can be separated from unrelated proteins by affinity chromatography using a conformation-dependent antibody provided herein such as AR3A.
  • a cross-neutralizing antibody such as, for example, AR3A, AR3B, AR3C or AR3D
  • the E2 polypeptide can be eluted at acidic, neutral or basic pH using: (1) 0.2M glycine pH 2.2, (2) 2M sodium thiocyanate (pH adjusted to pH 7.4 with 5OmM Tris-HCl); or (3) 0.2M glycine pH 11.5, and then further purified by size-exclusion chromatography.
  • the method provided herein for purifying E2 polypeptide allows for the purification of E2 polypeptides that properly fold to form the conformational epitope described herein.
  • the methods of purification using a conformation dependent antibody such as, but not limited to AR3A, provide for the purification of E2 polypeptide monomers that properly fold to form the E2 conformational epitope.
  • E2 polypeptides incorporated in a viral vector, for example, can be used as a nucleic acid-based vaccine to elicit an immune response against HCV.
  • the antibody is a cross-neutralizing antibody, i.e. one that neutralizes at least two HCV strains, isolates, species, quasispecies, subtypes or genotypes.
  • An antibody provided herein can be a polyclonal or monoclonal antibody.
  • Polyclonal antibodies can be obtained by immunizing a mammal with a mutant polypeptide provided herein, and then isolating antibodies from the blood of the mammal using standard techniques including, for example, enzyme linked immunosorbent assay (ELISA) to determine antibody titer and protein A chromatography to obtain the antibody-containing IgG fraction.
  • ELISA enzyme linked immunosorbent assay
  • a monoclonal antibody is a population of molecules having a common antigen binding site that binds specifically with a particular antigenic epitope.
  • a monoclonal antibody can be obtained by selecting an antibody-producing cell from a mammal that has been immunized with a mutant polypeptide provided herein and fusing the antibody-producing cell, e.g. a B cell, with a myeloma to generate an antibody- producing hybridoma.
  • a monoclonal antibody provided herein also can be obtained by screening a recombinant combinatorial library such as an antibody phage display library using, for example, a mutant polypeptide provided herein.
  • An immunologically-active fragment of an antibody is the biologically active fragment of an immunoglobulin molecule, for example, the F(ab) or F(ab') 2 fragment generated by cleavage of the antibody with an enzyme such as pepsin.
  • An antibody provided herein also can be a murine, chimeric, humanized or fully human antibody.
  • a murine antibody is an antibody derived entirely from a murine source, for example, an antibody derived from a murine hybridoma generated from the fusion of a mouse myeloma cell and a mouse B-lymphocyte cell.
  • a chimeric antibody is an antibody that has variable regions derived from a non-human source, e.g. murine or primate, and constant regions derived from a human source.
  • a humanized antibody has antigen-binding regions, e.g. complementarity-determining regions, derived from a mouse source, and the remaining variable regions and constant regions derived from a human source.
  • a fully human antibody is an antibody from human cells or derived from transgenic mice carrying human antibody genes.
  • a polyclonal antibody provided herein can be prepared by immunizing a suitable mammal with a mutant polypeptide provided herein.
  • the mammal can be, for example, a rabbit, goat, sheep, rabbit, hamster, cow, or mouse.
  • antibody molecules can be isolated from the mammal, e.g. from the blood or other fluid of the mammal, and further purified using standard techniques that include, without limitation, precipitation using ammonium sulfate, gel filtration chromatography, ion exchange chromatography or affinity chromatography using protein A.
  • an antibody-producing cell of the mammal can be isolated and used to prepare a hybridoma cell that secretes a monoclonal antibody provided herein.
  • Techniques for preparing monoclonal antibody-secreting hybridoma cells are known in the art. See, for example, Kohler and Milstein, Nature 256:495-97 (1975) and Kozbor etai, Immunol Today 4: 72 (1983).
  • a monoclonal antibody provided herein also can be prepared using other methods known in the art, such as, for example, expression from a recombinant DNA molecule, or screening of a recombinant combinatorial immunoglobulin library using a mutant polypeptide provided herein.
  • a chimeric antibody can be produced by expression from a nucleic acid that encodes a non-human variable region and a human constant region of .an antibody molecule. See, for example, Morrison et al., Proc. Nat. Acad. ScL U.S.A. 86: 6851 (1984).
  • a humanized antibody can be produced by expression from a nucleic acid that encodes non-human antigen-binding regions (complementarity-determining regions) and a human variable region (without antigen-binding regions) and human constant regions.
  • a mutant HCV E2 polypeptide or cross-neutralizing antibody provided herein can be used to detect the presence of HCV in a sample obtained from a subject, such as a mammal.
  • a diagnostic use is based on the detection of antibodies generated by a subject (e.g. a mammal) that has been infected with HCV.
  • Diagnostic use also can be based on detection of HCV antigens. Detection of an antibody-antigen complex indicates that the mammal has been exposed to or infected with HCV.
  • a mutant polypeptide provided herein can be used to detect the presence of anti-HCV antibodies in a sample from the mammal.
  • a cross-neutralizing antibody provided herein can be used to detect HCV particles or antigens in the sample.
  • the sample from the mammal can be a biological fluid such as blood or a cell or tissue sample.
  • the mutant E2 polypeptides or antibodies provided herein can be labeled with a detectable label.
  • the polypeptide or cross-neutralizing antibody provided herein can be labeled with a detectable molecule, which can be an enzyme such as, but not limited to, alkaline phosphatase, acetylcholinesterase, ⁇ - galactosidase or horseradish peroxidase; a prosthetic group such as, but not limited to, streptavidin, biotin, or avidin; a fluorescent group such as dansyl chloride, dichlorotriazinylamine, dichlorotriazinylamine fluorescein, fluorescein, fluorescein isothiocyanate, phycoerythrin, rhodamine, umbelliferone; a luminescent group such as luminal; a bioluminescent group such as a
  • a polypeptide provided herein can be used to generate cross-neutralizing antibodies against HCV.
  • a polypeptide provided herein can be used to elicit an immune response in a subject, such as a mammal.
  • Antibodies that bind specifically with the mutant E2 polypeptide provided herein can be isolated using known methods as described above.
  • a mutant polypeptide provided herein is particularly useful to focus the immune response to the conserved AR3 neutralizing epitopes as the immunogenicity of the hypervariable regions and the ARl residues are dampened by deletion of a large portion of the hypervariable region and substitution of important selected ARl residues.
  • kits for eliciting an immune response in a subject comprising administering to the subject a mutant E2 polypeptide provided herein and then isolating antibodies or antibody producing cells from the subject using methods known to those of skilled in the art.
  • the subject can be a rabbit, rat, mouse, sheep, cow, monkey, horse, goat or a pig.
  • the method is particularly useful to generate antibodies against conserved HCV epitopes.
  • the method can be used to develop passive vaccines containing one or more anti-HCV antibodies provided herein.
  • a polypeptide provided herein also can be used to screen for anti-HCV agents, such as those that block viral entry into target cells.
  • an E2 polypeptide provided herein can be used to screen for agents that bind to an E2 polypeptide provided herein and prevent binding of the E2 polypeptide with a cell receptor.
  • a polypeptide or cross-neutralizing antibody provided herein can be used to prevent or treat a new or recurring HCV infection, or prevent or reduce HCV replication, as well as treat the associated disease condition or clinical symptoms.
  • the mutant HCV polypeptide provided herein can be used to provide immune protection against HCV.
  • the immune protection against HCV provided by the immunogenic polypeptide can be any immune response, cellular or humoral, that either inhibits or helps to prevent HCV infection.
  • the immunogenic polypeptide of the present invention can bind to CD81, induce antibodies associated with resolving HCV infection, induce production of cytokines, induce antibodies that can neutralize HCV binding to host cells, or prime an immune system against secondary HCV infection or exposure.
  • the immune protection provided by the immunogenic polypeptide of the present invention can be protective against more than one of the HCV genotypes.
  • HCV infection or replication is indicated by the amount of HCV particles or the amount of HCV RNA in a sample from the subject determined using methods known in the art and also those described herein.
  • HCV infection is also indicated by clinical symptoms described further below.
  • the E2 polypeptide provided herein, corresponding nucleic acid or cross- neutralizing antibody provided herein can be used to prevent or reduce transmission, to prevent or treat disease progression, and to prevent or reduce HCV replication or reduce viral load.
  • Treatment includes the alleviation or diminishment of at least one symptom typically associated with the infection.
  • the treatment cures, e.g., substantially inhibits viral infection and/or eliminates the symptoms associated with the infection.
  • Symptoms of HCV exposure or infection include, without limitation, inflammation of the liver, decreased appetite, fatigue, abdominal pain, jaundice, flu- like symptoms, itching, muscle pain, joint pain, intermittent low-grade fevers, sleep disturbances, nausea, dyspepsia, cognitive changes, depression headaches and mood changes.
  • Subjects that can benefit from the polypeptide, nucleic acid or antibody provided herein can be identified using the diagnostic and screening techniques discussed above.
  • HCV infection can be diagnosed by detecting antibodies to the virus using the mutant E2 polypeptide provided herein, detecting the HCV itself using a cross-neutralizing antibody provided herein, detecting liver inflammation by biopsy, liver cirrhosis, portal hypertension, thyroiditis, cryoglobulinemia and glomerulonephritis.
  • diagnosis of exposure or infection or identification of one who is at risk of exposure to HCV can be based on medical history, abnormal liver enzymes or liver function tests during routine blood testing.
  • infection can be diagnosed using polymerase chain reaction (PCR) for detecting viral nucleic acids, enzyme linked immunosorbent assay (ELISA) for detecting viral antigens or anti-viral antibodies, and immunofluorescence for detecting viral antigens.
  • PCR polymerase chain reaction
  • ELISA enzyme linked immunosorbent assay
  • a polypeptide or antibody provided herein can be combined with an appropriate sample from the patient to produce a complex.
  • the complex in turn can be detected with a marker reagent for binding with such a complex.
  • Typical marker reagents include secondary antibodies selective for the complex, secondary antibodies selective for certain epitopes of the polypeptide or antibody or a label attached to the polypeptide or antibody itself.
  • radioimmunoassay RIA
  • radioallergosorbent test RAST
  • radioimmunosorbent test RIST
  • immunoradiometric assay IRMA
  • Fair assay fluorescence immunoassay
  • FOA fluorescence immunoassay
  • ELISA enzyme linked immunosorbent assay
  • Labels including radioactive labels, chemical labels, fluorescent labels, luciferase and the like also can be directly attached to the polypeptide according to the techniques described in U.S. Patent No. (BN patent cite), the disclosure of which is incorporated herein by reference.
  • a subject that can benefit from a polypeptide, nucleic acid or cross-neutralizing antibody provided herein includes one who is likely to be or has been exposed to HCV.
  • exemplary subjects include, without limitation, someone present in an area where HCV is prevalent or commonly transmitted, e.g., Africa, Southeast Asia, China, South Asia, Australia, India, the United States, Russia, as well as Central and South American countries.
  • a subject who is likely to be or has been exposed to HCV also includes a recipient of donated body tissues or fluids including, for example, a recipient of blood or one or more of its components such as plasma, platelets, or stem cells and an organ or cell transplant recipient such as a liver transplantee.
  • a subject who is likely to be or has been exposed to HCV also can include medical, clinical or dental personnel handling body tissues and fluids.
  • a subject e.g., a mammal
  • who has been exposed to HCV includes, without limitation, someone who has had contact with the body tissue or fluid, e.g. blood, of an infected person or otherwise have come in contact with HCV.
  • a subject e.g., a mammal
  • who can benefit from a polypeptide or cross-neutralizing antibody provided herein includes one who is susceptible to HCV infection or one who has recurring HCV infection.
  • kits for preventing a new or recurring HCV infection and its associated symptoms and/or complications such as by preventing or reducing HCV replication in a subject (e.g. a mammal) infected with HCV.
  • a polypeptide, nucleic acid or cross-neutralizing antibody provided herein can be used prophylactically to prevent a susceptible individual from being infected with HCV or to prevent recurring HCV infection, for example, in an individual who has received a liver transplant.
  • a polypeptide or cross-neutralizing antibody provided herein can be used to prevent or treat infection of a cell, e.g. a mammalian cell, such as a human cell.
  • a polypeptide, nucleic acid or cross-neutralizing antibody provided herein can be used to prevent or treat acute or chronic HCV infection, or prevent or reduce HCV replication, in a subject, e.g. a mammal such as a human.
  • an E2 polypeptide or a nucleic acid encoding an E2 polypeptide provided herein can be used as an active vaccine, a nucleic acid or DNA-based vaccine, or be incorporated into vaccine carriers, to elicit a protective immune response in a subject.
  • Exemplary vaccines include vaccines that are effective for the prevention of HCV infection by one or more HCV genotypes.
  • Methods of preventing or treating HCV infection include contacting a cell with an effective amount of an antibody provided herein; mixing biological fluids, cells or tissues to be administered or transplanted into a subject with a polypeptide, nucleic acid or antibody provided herein prior to the administration or transplant; or administering to a subject such as a human a therapeutically effective amount of a polypeptide, nucleic acid or antibody provided herein.
  • in vitro methods of preventing HCV infection or transmission by contacting biological samples such as fluids, cells or tissues containing the virus with an effective amount of the polypeptide, nucleic acid or antibody provided herein, as well as in vivo methods of treating or preventing HCV infection by administering the polypeptide, nucleic acid or antibody to the subject.
  • a polypeptide, nucleic acid or antibody provided herein can be administered in a variety of ways.
  • Routes of administration include, without limitation, oral, parenteral (including subcutaneous, intravenous, intramuscular and intraperitoneal), rectal, vaginal, dermal, transdermal (topical), transmucosal, intrathoracic, intrapulmonary and intranasal (respiratory) routes.
  • the means of administration can be by injection, using a pump or any other appropriate mechanism.
  • a polypeptide, nucleic acid or antibody provided herein can be administered in a single dose, in multiple doses, in a continuous or intermittent manner, depending, for example, upon the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of the polypeptide, nucleic acid or antibody provided herein can be essentially continuous over a pre-selected period of time or can be in a series of spaced doses.
  • methods of eliciting an immune response in a subject e.g.
  • a mammal that involves administering a modified polypeptide, nucleic acid or antibody provided herein at a select time and then administering a second, third, fourth or additional doses at select times after the first administration. Both local and systemic administrations are contemplated.
  • the dosage to be administered to a subject can be any amount appropriate to reduce or prevent viral infection or to treat at least one symptom associated with the viral infection.
  • a subject e.g. a mammal
  • dosages can be any amount appropriate to reduce or prevent viral infection or to treat at least one symptom associated with the viral infection.
  • Some factors that determine appropriate dosages are well known to those of ordinary skill in the art and can be addressed with routine experimentation. For example, determination of the physicochemical, toxicological and pharmacokinetic properties can be made using standard chemical and biological assays and through the use of mathematical modeling techniques known in the chemical, pharmacological and toxicological arts. The therapeutic utility and dosing regimen can be extrapolated from the results of such techniques and through the use of appropriate pharmacokinetic and/or pharmacodynamic models.
  • the dosage will also depend on the polypeptide or antibody chosen and whether prevention or treatment is to be achieved, and if the polypeptide or antibody is chemically mutant. Such factors can be readily determined by the clinician employing viral infection models such as in vitro HCV infection system described herein, or other animal models or test systems that are available in the art. The precise amount to be administered to a subject (e.g. a mammal) such as a human will be the responsibility of the attendant physician. The amount useful to establish treatment of HCV can be determined by diagnostic and therapeutic techniques well known to those of ordinary skill in the art.
  • the dosage can be determined by titrating a sample of the patient's blood sera with the polypeptide or antibody to determine the end point beyond which no further immunocomplex is formed. Such titrations can be accomplished by the diagnostic techniques discussed below. Available dosages include administration of from about 1 to about 1 million effective units of antibody per day, wherein a unit is that amount of polypeptide, which will provide at least 1 microgram of antigen-polypeptide complex. In some examples, about 10 to about 100,000 units of antibody per day can be administered.
  • one or more mutant polypeptides or antibody provided herein can be administered as single or divided dosages, for example, of at least about 0.01 mg/kg to about 500, 750 or 1000 mg/kg, of at least about 0.01 mg/kg to about 300 to 500 mg/kg, at least about 0.1 mg/kg to about 100 to 300 mg/kg or at least about 1 mg/kg to about 50 to 100 mg/kg of body weight, although other dosages can provide beneficial results.
  • the one or more polypeptide or antibody provided herein can be administered as soon as possible, e.g. within 24 hours if possible, after exposure to HCV. To prevent recurrent HCV infection, e.g.
  • a mutant polypeptide or antibody provided herein can be administered prior to and after transplantation.
  • the polypeptide or antibody provided herein can be administered during the anhepatic phase, as well as during the post-operative phase.
  • the polypeptide, nucleic acid or antibody provided herein can be administered daily, biweekly or monthly after the transplant.
  • the polypeptide, nucleic acid or antibody provided herein can be administered daily for the first week after transplant, weekly for two, three or more weeks after the transplant and then monthly.
  • the absolute weight of a polypeptide or antibody included in a unit dose can vary widely.
  • the unit dosage can vary from about 0.01 g to about 50 g, from about 0.01 g to about 35 g, from about 0.1 g to about 25 g, from about 0.5 g to about 12 g, from about 0.5 g to about 8 g, from about 0.5 g to about 4 g, or from about 0.5 g to about 2 g.
  • the daily dose of a polypeptide, nucleic acid or antibody provided herein can vary as well. Such daily doses can range, for example, from about 0.1 g/day to about 50 g/day, from about 0.1 g/day to about 25 g/day, from about 0.1 g/day to about 12 g/day, from about 0.5 g/day to about 8 g/day, from about 0.5 g/day to about 4 g/day, and from about 0.5 g/day to about 2 g/day.
  • a polypeptide, nucleic acid or antibody provided herein can be used alone or in combination with a second medicament.
  • the second medicament can be another polypeptide or antibody provided herein, a known antiviral agent such as, for example, an interferon-based therapeutic or another type of antiviral medicament such as ribavirin.
  • a polypeptide, nucleic acid or antibody provided herein also can be used in combination with one or more agents to enhance the immune response in a subject.
  • the polypeptide, nucleic acid or antibody provided herein can be administered in combination with other therapeutic agents including, without limitation, immunoregulatory agents, immunoglobulin, cytokines, lymphokines, and chemokines, e.g., IL-2, modified IL-2 (e.g. Cl 25S), GM-CSF, IL-12, gamma- interferon, IP-IO, MIPl ⁇ , or RANTES.
  • the second medicament also can be an anticancer, antibacterial, or another antiviral agent.
  • the antiviral agent can act at any step in the life cycle of the virus from initial attachment and entry to egress.
  • the second antiviral agent can interfere with attachment, fusion, entry, trafficking, translation, viral polyprotein processing, viral genome replication, viral particle assembly, egress or budding.
  • the antiviral agent can be an attachment inhibitor, entry inhibitor, a fusion inhibitor, a trafficking inhibitor, a replication inhibitor, a translation inhibitor, a protein processing inhibitor, an egress inhibitor, in essence an inhibitor of any viral function.
  • the effective amount of the second medicament will follow the recommendations of the manufacturer of the second medicament, as well as the judgment of the attending physician, and will be guided by the protocols and administrative factors for amounts and dosing as indicated in the PHYSICIAN'S DESK REFERENCE.
  • the polypeptide, nucleic acid or antibody provided herein can be administered in combination with an adjuvant.
  • the polypeptide, nucleic acid or antibody provided herein can be administered with any suitable adjuvant for stimulating immune response, e.g., providing immune protection.
  • it can be a particulate or a non-particulate adjuvant.
  • a particulate adjuvant usually includes, without limitation, aluminum salts, calcium salts, water-in-oil emulsions, oil-in water emulsion, immune stimulating complexes (ISCOMS) and ISCOM matrices (U.S. Pat. No.
  • a non-particulate adjuvant usually includes, without limitation, muramyl dipeptide (MDP) and derivatives, e.g., treonyl MDP or murametide, non-ionic block copolymers, saponins, e.g., Quil A and QS21, lipid A or its derivative 4' monophosphoryl lipid A (MPL), trehalose dimycolate (TDM), various cytokines including gamma-interferon and interleukins 2 or 4, carbohydrate polymers, derivatized polysaccharides, e.g., di ethyl aminoethyl dextran, and bacterial toxins, e.g., cholera toxin or E. coli labile toxin.
  • the polypeptide e.g., treonyl MDP or murametide, non-ionic block copolymers
  • saponins e.g., Quil A and QS21
  • MPL
  • nucleic acid or antibody provided herein for inhibition and treatment of HCV infection, methods available in the art and those described herein can be used.
  • the effectiveness of the method of treatment can be assessed by monitoring the patient for signs or symptoms of the viral infection as discussed above, as well as determining the presence and/or amount of viral particle or viral RNA present in the blood, e.g. the viral load, using methods known in the art.
  • Viral infection or replication in the presence or absence of a polypeptide or antibody provided herein can be evaluated, for example, by determining intracellular viral RNA levels or the number of viral foci by immunoassays using antibodies against viral proteins as described herein.
  • a polypeptide or antibody is effective for treatment and inhibition of HCV if it can inhibit or reduce viral infection or replication by any amount, for example, by 2 fold or more than 2 fold.
  • a polypeptide or antibody provided herein can inhibit or reduce HCV infection by 2-5 folds, 5-10 folds, or more than 10 folds.
  • a polypeptide, nucleic acid or antibody provided herein also can be used to increase the safety of blood and blood products, to increase the safety of clinical laboratory samples and to increase the safety of biological tissues as well as articles, devices, or instruments intended for preventative or therapeutic use.
  • a polypeptide, nucleic acid or antibody provided herein can be added to blood or blood products such as plasma, platelets, and blood or marrow cells prior to use.
  • a polypeptide, nucleic acid or antibody provided herein can be combined with cells or tissues prior to use or administration. It can be coated on articles, devices or instruments such as, for example, valves, bags and stents.
  • purified preparations containing a mutant polypeptide provided herein or a preparation containing a cross-neutralizing antibody provided herein are provided herein.
  • a purified preparation of a mutant polypeptide provided herein at least 50 % of the mutant polypeptides in the preparation are folded in a conformation such that the discontinuous epitopes (i.e. amino acid segments corresponding to amino acids 412 to 424, amino acids 436 to 447 and amino acids 523 to 540 of HCV strain H77) come together to form a conformational epitope that can bind with a conformation-dependent antibody such as a cross-neutralizing antibody, for example, AR3A, AR3B, AR3C or AR3D.
  • a conformation-dependent antibody such as a cross-neutralizing antibody
  • polypeptide preparation at least about 50 %, 55 %, 60 %, 65 %, 70 %, 75 %, 80 %, 85 %, 90 % or 95 % of the mutant polypeptides are folded as described above.
  • mutant polypeptides are folded as described.
  • a larger proportion of the antibodies are cross-neutralizing antibodies.
  • an antibody preparation can be a biological sample such as blood or plasma obtained from a subject (e.g. a mammal) that has been immunized with a mutant polypeptide provided herein.
  • the blood sample contains a larger proportion of cross- neutralizing antibodies than a blood sample obtained from a similar animal that has been immunized with a naturally-occurring E2 polypeptide.
  • Such a cross-neutralizing antibody preparation can be a partially purified or purified polypeptide preparation, i.e. a preparation resulting from one or more protein purification steps known in the art as well as those discussed herein.
  • Such cross- neutralizing antibody preparation contains at least about 2 %, 5%, 10 %, 20 %, 30 %, 40 %, 50 %, 55 %, 60 %, 65 %, 70 %, 75 %, or 80 % cross- neutralizing anti-HCV antibodies.
  • such cross-neutralizing antibody preparation can contain about 5 %, 6 %, 7 %, 8 %, 9 %, 10 %, 12 %, 13 %, 14 %, 15 %, 16 %, 17 %, 18 %, 19 %, 20 %, 22 %, 23 %, 24 %, 25 %, 26 %, 27 %, 28 %, 29 %, 30 %, 32 %, 33 %, 34 %, 35 %, 36 %, 37 %, 38 %, 39 %, 40 %, 42 %, 43 %, 44 %, 45 %, 46 %, 47 %, 48 %, 49 %, 50 %, 52 %, 53 %, 54 %, 55 %, 56 %, 57 %, 58 %, 59 %, 60 %, 62 %, 63 %, 64 %, 65 %, 66 %, 67 %, 68
  • mutant polypeptides and cross-neutralizing antibodies provided herein are described above. Preparations of these can be obtained using protein purification procedures known to those skilled in the art. See, for example, CURRENT PROTOCOLS IN PROTEIN SCIENCE, Coligan et al, eds., John Wiley & Sons, Inc., 1997.
  • compositions comprising a mutant polypeptide, nucleic acid or antibody provided herein.
  • a mutant polypeptide or antibody provided herein is obtained, e.g. by expression in a host cell or using polymerase chain reaction, purified as necessary or desired and then lyophilized and stabilized.
  • the polypeptide, nucleic acid or antibody can then be adjusted to the appropriate concentration and then combined with other agent(s) or pharmaceutically acceptable carrier(s).
  • a pharmaceutical formulation containing therapeutic amounts of one or more polypeptides, nucleic acids or antibodies provided herein can be prepared by procedures known in the art using well-known and readily available ingredients.
  • one or more polypeptides, nucleic acids or antibodies can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, solutions, suspensions, powders, aerosols and the like.
  • excipients, diluents, and carriers that are suitable for such formulations include buffers, as well as fillers and extenders such as starch, cellulose, sugars, mannitol, and silicic derivatives.
  • Binding agents also can be included such as carboxymethyl cellulose, hydroxymethylcellulose, hydroxypropyl methylcellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl-pyrrolidone.
  • Moisturizing agents can be included such as glycerol, disintegrating agents such as calcium carbonate and sodium bicarbonate. Agents for retarding dissolution also can be included such as paraffin. Resorption accelerators such as quaternary ammonium compounds also can be included. Surface active agents such as cetyl alcohol and glycerol monostearate can be included. Adsorptive carriers such as kaolin and bentonite can be added. Lubricants such as talc, calcium and magnesium stearate, and solid polyethyl glycols also can be included. Preservatives also can be added. The compositions provided herein also can contain thickening agents such as cellulose and/or cellulose derivatives.
  • polypeptides, nucleic acids or antibodies can be present as a powder, a granular formulation, a solution, a suspension, an emulsion or in a natural or synthetic polymer or resin for ingestion of the active ingredients from a chewing gum.
  • the active polypeptide also can be presented as a bolus, electuary or paste.
  • the formulations can, where appropriate, be conveniently presented in discrete unit dosage forms and can be prepared by any of the methods well known to the pharmaceutical arts including the step of mixing the therapeutic agent with liquid carriers, solid matrices, semi-solid carriers, finely divided solid carriers or combinations thereof, and then, if necessary, introducing or shaping the product into the desired delivery system.
  • the total active ingredients in such formulations comprise from 0.1 to 99.9% by weight of the formulation.
  • One or more polypeptides, nucleic acids or antibodies provided herein also can be formulated as elixirs or solutions for convenient oral administration or as solutions appropriate for parenteral administration, for instance by intramuscular, subcutaneous, intraperitoneal or intravenous routes.
  • a pharmaceutical formulation containing one or more therapeutic polypeptides, nucleic acids or antibodies provided herein also can take the form of an aqueous or anhydrous solution or dispersion, or alternatively the form of an emulsion or suspension or salve.
  • polypeptides, nucleic acids or antibodies can be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and can be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion containers or in multi-dose containers. As noted above, preservatives can be added to help maintain the shelf life of the dosage form.
  • the polypeptides, nucleic acids or antibodies and other ingredients can form suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • polypeptides, nucleic acids or antibodies and other ingredients can be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water
  • formulations can contain pharmaceutically acceptable carriers, vehicles and adjuvants that are well known in the art. It is possible, for example, to prepare solutions using one or more organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name "Dowanol,” polyglycols and polyethylene glycols, C1-C4 alkyl esters of short-chain acids, ethyl or isopropyl lactate, fatty acid triglycerides such as the products marketed under the name "Miglyol,” isopropyl myristate, animal, mineral and vegetable oils and polysiloxanes.
  • organic solvent(s) that is/are acceptable from the physiological standpoint, chosen, in addition to water, from solvents such as acetone, ethanol, isopropyl alcohol, glycol ethers such as the products sold under the name "Dowanol,” polygly
  • antioxidants chosen from antioxidants, surfactants, other preservatives, film-forming, keratolytic or comedolytic agents, perfumes, flavorings and colorings.
  • Antioxidants such as t-butylhydroquinone, butylated hydroxyanisole, butylated hydroxytoluene and ⁇ -tocopherol and its derivatives can be added.
  • the one or more polypeptides, nucleic acids or antibodies are formulated as a microbicide, which is administered topically or to mucosal surfaces such as the vagina, the rectum, eyes, nose and the mouth.
  • the therapeutic agents can be formulated as is known in the art for direct application to a target area.
  • Forms chiefly conditioned for topical application take the form, for example, of creams, milks, gels, dispersion or microemulsions, lotions thickened to a greater or lesser extent, impregnated pads, ointments or sticks, aerosol formulations (e.g., sprays or foams), soaps, detergents, lotions or cakes of soap.
  • an agent provided herein can be formulated as a vaginal cream or a microbicide to be applied topically.
  • Other conventional forms for this purpose include wound dressings, coated bandages or other polymer coverings, ointments, creams, lotions, pastes, jellies, sprays, and aerosols.
  • the one or more polypeptides, nucleic acids or antibodies provided herein can be delivered via patches or bandages for dermal administration.
  • the polypeptides, nucleic acids or antibodies can be formulated to be part of an adhesive polymer, such as polyacrylate or acryl ate/vinyl acetate copolymer.
  • the backing layer can be any appropriate thickness that will provide the desired protective and support functions.
  • a suitable thickness will generally be from about 10 to about 200 microns.
  • Ointments and creams can, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions can be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • the active agents also can be delivered via iontophoresis, e.g., as disclosed in U.S. Patent Nos. 4,140,122; 4,383,529; or 4,051 ,842.
  • the percent by weight of one or more polypeptides, nucleic acids or antibodies provided herein present in a topical formulation will depend on various factors, but generally will be from 0.01 % to 95 % of the total weight of the formulation, and typically 0.1-85 % by weight.
  • Drops such as eye drops or nose drops, can be formulated with one or more of the polypeptides, nucleic acids or antibodies in an aqueous or non-aqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents.
  • Liquid sprays are conveniently delivered from pressurized packs. Drops can be delivered via a simple eye dropper-capped bottle, or via a plastic bottle adapted to deliver liquid contents dropwise, via a specially shaped closure.
  • the one or more polypeptides, nucleic acids or antibodies further can be formulated for topical administration in the mouth or throat.
  • the active ingredients can be formulated as a lozenge further comprising a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the composition in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the composition provided herein in a suitable liquid carrier.
  • the pharmaceutical formulations provided herein can include, as optional ingredients, pharmaceutically acceptable carriers, diluents, solubilizing or emulsifying agents, and salts of the type that are available in the art.
  • pharmaceutically acceptable carriers such as physiologically buffered saline solutions and water.
  • diluents such as phosphate buffered saline solutions pH 7.0-8.0.
  • polypeptides, nucleic acids or antibodies provided herein also can be administered to the respiratory tract.
  • aerosol pharmaceutical formulations and dosage torms tor use in the methods provided herein comprise an amount of at least one of the polypeptides, nucleic acids or antibodies provided herein effective to treat or prevent the clinical symptoms of the viral infection. Any statistically significant attenuation of one or more symptoms of the infection that has been treated pursuant to the method provided herein is considered to be a treatment of such infection within the scope provided herein.
  • the composition can take the form of a dry powder, for example, a powder mix of one or more polypeptides, nucleic acids or antibodies and a suitable powder base such as lactose or starch.
  • the powder composition can be presented in unit dosage form in, for example, capsules or cartridges, or, e.g., gelatin or blister packs from which the powder can be administered with the aid of an inhalator, insufflator, or a metered-dose inhaler (see, for example, the pressurized metered dose inhaler (MDI) and the dry powder inhaler disclosed in Newman, S. P. in Aerosols and the Lung. Clarke. S. W. and Davia, D. eds., pp. 197-224, Butterworths, London, England, 1984).
  • MDI pressurized metered dose inhaler
  • the dry powder inhaler disclosed in Newman, S. P. in Aerosols and the Lung. Clarke. S. W. and Davia, D. e
  • the one or more polypeptides, nucleic acids or antibodies provided herein also can be administered in an aqueous solution when administered in an aerosol or inhaled form.
  • other aerosol pharmaceutical formulations can comprise, for example, a physiologically acceptable buffered saline solution containing between about 0.1 mg/mL and about 100 mg/mL of one or more of the polypeptides, nucleic acids or antibodies provided herein specific for the indication or disease to be treated.
  • Dry aerosol in the form of finely divided solid polypeptide, nucleic acid or antibody particles that are not dissolved or suspended in a liquid are also useful in the practice provided herein.
  • Polypeptides, nucleic acids or antibodies provided herein can be formulated as dusting powders and comprise finely divided particles having an average particle size of between about 1 and 5 ⁇ m, alternatively between 2 and 3 ⁇ m.
  • Finely divided particles can be prepared by pulverization and screen filtration using techniques well known in the art.
  • the particles can be administered by inhaling a predetermined quantity of the finely divided material, which can be in the form of a powder. It will be appreciated that the unit content of active ingredient or ingredients contained in an individual aerosol dose of each dosage form need not in itself constitute an effective amount for treating the particular infection, indication or disease since the necessary effective amount can be reached by administration of a plurality of dosage units.
  • the effective amount can be achieved using less than the dose in the dosage form, either individually, or in a series of administrations.
  • the one or more polypeptides, nucleic acids or antibodies provided herein are conveniently delivered from a nebulizer or a pressurized pack or other convenient means of delivering an aerosol spray.
  • Pressurized packs can comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit can be determined by providing a valve to deliver a metered amount.
  • Nebulizers include, but are not limited to, those described in U.S. Patent Nos. 4,624,251; 3,703,173; 3,561,444; and 4,635,627. Aerosol delivery systems of the type disclosed herein are available from numerous commercial sources including Fisons Corporation (Bedford, Mass.), Schering Corp. (Kenilworth, NJ) and American Pharmoseal Co., (Valencia, CA). For intra-nasal administration, the therapeutic agent also can be administered via nose drops, a liquid spray, such as via a plastic bottle atomizer or metered-dose inhaler. Typical of atomizers are the Mistometer (Wintrop) and the Medihaler (Riker).
  • An exemplary formulation involves lyophilized polypeptides, nucleic acids or antibodies and separate pharmaceutical carrier. Immediately prior to administration, the formulation is constituted by combining the lyophilized polypeptides, nucleic acids or antibodies and pharmaceutical carrier. Administration by a parenteral or oral regimen will deliver the polypeptides, nucleic acids or antibodies to the desired site of action.
  • Pharmaceutical formulations of the polypeptides, nucleic acids or antibodies provided herein can be prepared as liquids, gels and suspensions. Exemplary formulations are suitable for injection, insertion or inhalation. Injection can be accomplished by needle, cannula, catheter and the like. Insertion can be accomplished by lavage, trochar, spiking, surgical placement and the like.
  • Inhalation can be accomplished by aerosol, spray or mist formulation.
  • the polypeptides, nucleic acids or antibodies provided herein also can be administered topically such as to the epidermis, the buccal cavity and instillation into the ear, eye and nose.
  • the polypeptides, nucleic acids or antibodies can be present in the pharmaceutical formulation at concentrations ranging from about 1 percent to about 50 percent, such as about 1 percent to about 20 percent, such as, for example, about 2 percent to about 10 percent by weight relative to the total weight of the formulation.
  • a polypeptide, nucleic acid or antibody provided herein also can be used in combination with one or more known therapeutic agents, for example, a pain reliever; an antiviral agent such as an anti-HBV, other anti-HCV (HCV inhibitor, HCV protease inhibitor) or an anti-herpetic agent; an antibacterial agent; an anti-cancer agent; an anti-inflammatory agent; an antihistamine; a bronchodilator; an immunomodulatory agent; and appropriate combinations thereof, whether for the conditions described or some other condition.
  • a pain reliever an antiviral agent such as an anti-HBV, other anti-HCV (HCV inhibitor, HCV protease inhibitor) or an anti-herpetic agent
  • an antibacterial agent such as an anti-cancer agent
  • an anti-inflammatory agent such as an antihistamine; a bronchodilator; an immunomodulatory agent; and appropriate combinations thereof, whether for the conditions described or some other condition.
  • articles of manufacture that include a pharmaceutical composition containing one or more polypeptides, nucleic acids or antibodies provided herein for controlling microbial infections.
  • Such articles can be a useful device such as a vaginal ring, a condom, a bandage or a similar device.
  • the device holds a therapeutically effective amount of a pharmaceutical composition for controlling viral infections.
  • the device can be packaged in a kit along with instructions for using the pharmaceutical composition for control of the infection.
  • the pharmaceutical composition includes at least one polypeptide, nucleic acid or antibody provided herein, in a therapeutically effective amount such that viral infection is controlled.
  • An article of manufacture also can be a vessel or filtration unit that can be used for collection, processing or storage of a biological sample containing a polypeptide or antibody provided herein.
  • the vessel can be evacuated.
  • Vessels include, without limitation, a capillary tube, a vacutainer, a collection bag for blood or other body fluids, a cannula, a catheter.
  • the filtration unit can be part of another device, for example, a catheter for collection of biological fluids.
  • the one or more polypeptides or antibodies provided herein also can be adsorbed onto or covalently attached to the article of manufacture, for example, a vessel or filtration unit.
  • the material in the article when material in the article is decanted therefrom or passed through, the material will not retain substantial amounts of the polypeptides or antibodies.
  • Adsorption or covalent attachment of the one or more polypeptides or antibodies to the article kills viruses or prevents their transmission, thereby helping to control viral infection.
  • the one or more polypeptides or antibodies provided herein can be in filtration units integrated into biological collection catheters and vials, or added to collection vessels to remove or inactivate viral particles that can be present in the biological samples collected, thereby preventing transmission of the disease.
  • compositions comprising one or more polypeptides, nucleic acids or antibodies provided herein and one or more clinically useful agents such as a biological stabilizer.
  • Biological stabilizer includes, without limitation, an anticoagulant, a preservative and a protease inhibitor.
  • Anticoagulants include, without limitation, oxalate, ethylene diamine tetraacetic acid, citrate and heparin.
  • Preservatives include, without limitation, boric acid, sodium formate and sodium borate.
  • Protease inhibitors include inhibitors of dipeptidyl peptidase IV.
  • compositions comprising an agent provided herein and a biological stabilizer can be included in a collection vessel such as a capillary tube, a vacutainer, a collection bag for blood or other body fluids, a cannula, a catheter or any other container or vessel used for the collection, processing or storage of biological samples.
  • a collection vessel such as a capillary tube, a vacutainer, a collection bag for blood or other body fluids, a cannula, a catheter or any other container or vessel used for the collection, processing or storage of biological samples.
  • compositions comprising one or more polypeptides, nucleic acids or antibodies provided herein and a biological sample such as blood, semen or other body fluids that is to be analyzed in a laboratory or introduced into a recipient subject (e.g. a mammal).
  • a biological sample such as blood, semen or other body fluids that is to be analyzed in a laboratory or introduced into a recipient subject (e.g. a mammal).
  • one or more polypeptides, nucleic acids or antibodies provided herein can be mixed with blood prior to laboratory processing and/or transfusions.
  • the one or more polypeptides, nucleic acids or antibodies are present in at least about 0.15 mg/mL of the sample, e.g.
  • the one or more polypeptides, nucleic acids or antibodies provided herein can be included in physiological media used to store and transport biological tissues, including transplantation tissues.
  • physiological media used to store and transport biological tissues including transplantation tissues.
  • liver, heart, kidney and other tissues can be bathed in media containing the present agents to inhibit viral transmission to transplant recipients.
  • the one or more polypeptides, nucleic acids or antibodies are present in at least about 1.5 mg/kg of the sample, e.g.
  • Huh-7 (Zhong, J. et al., Proc. Natl. Acad. ScL U.S.A. 102, 9294-9299 (2005)) and 293T cells were grown in Dulbecco's Modified Eagle Medium (D-MEM) supplemented with 10% fetal calf serum (FCS) (Invitrogen).
  • D-MEM Dulbecco's Modified Eagle Medium
  • FCS fetal calf serum
  • bone marrow mononuclear cell RNA from a 35-year-old female patient with Sjogren's syndrome and chronic HCV infection was used as source material for an IgGl Fab phage display library (Maruyama, T. etal, Am. J. Pathol. 165, 53-61 (2004)).
  • the donor was diagnosed with HCV in 1991 and developed mixed cryoglobulinemia, symptoms of Sjogren's syndrome and tested positive for antinuclear antibody in 1994.
  • the donor was treated with interferon- ⁇ with initial decrease in viral load but the treatment was stopped due to severe drop in platelet count (idiopathic thrombocytopenic purpura).
  • Bone marrow samples were collected for the evaluation of neutropenia as an outpatient clinical procedure at Scripps Clinic. After meeting the needs of clinical pathology, a fraction of the biopsy was used to construct the antibody library. The human subjects protocol was approved by the Human Subjects Committee for General Clinical Research Center of Scripps Clinic and informed consent was obtained from the donor. Due to subsequent relapse of HCV, the donor underwent a liver transplant in 2000 and has been maintained on anti-rejection medications since. The viral genotype in this donor was not determined at the time of tissue donation but was found to be genotype 1 a seven years later.
  • the Fab heavy chains were expressed as a fusion protein with the phage gene HI surface protein for display.
  • the library was amplified in XL-I Blue cells (Stratagene) using 0.3% SeaPrep agarose (BioWhittaker) in SuperBroth (SB) Medium by a semisolid phase amplification method.
  • the phagemid library was transformed into E. coli (XL-I Blue) (Stratagene) by electroporation and the phage was propagated overnight with VCS-Ml 3 helper phage (Stratagene).
  • Recombinant E2 glycoprotein (genotype Ia, amino acids 388- 644; Lesniewski, R. et al., J. Med. Virol. 45, 415-422 (1995)) was coated directly onto a microtiter plate overnight at 4 0 C (Costar). The wells were washed and then blocked with 4% non-fat dry milk in phosphate-buffered saline (PBS).
  • the phage library was added to the wells and incubated for 1-2 hours at 37°C and unbound phage washed away with PBS. Bound phage were eluted and used to infect freshly grown E. coli (XLl -Blue) (Stratagene) for titration on LB agar plates with carbenicillin. The phage libraries were panned for four consecutive rounds with increasing washing stringency.
  • Library panning by an epitope masking strategy was repeated using recombinant E1E2 fused to glutathione S transferase (GST-E1E2; Chan-Fook, C. et al., Virology TTb, 60-66 (2000)) pre-incubated with Fabs obtained above.
  • GST-E1E2 was first captured with goat anti-GST antibody (Amersham Biosciences) and the wells were washed and blocked with 4% non-fat dry milk in PBS.
  • Fabs obtained from the panning using E2 antigen above were added to the captured antigens to mask corresponding specific epitopes.
  • the epitope-masked GST-El E2 was used to pan the phage library as described above. It is important to note that, highly isolate-specific antibodies, e.g. those against HVRl, were not selected due to the use of heterologous antigens in panning. Screening of Fab displayed phage. Single individual colonies were isolated from titration plates after the 2nd, the
  • Vector plgGl is a derivative of pDR12 in which heavy and light chain cloning sites were altered to XhollBstEll and SacllXbal sites to facilitate direct cloning of the antibody gene fragments.
  • the heavy and light chain genes of Fab Cl were amplified by PCR then inserted sequentially into the SacVXbal and Hindlll/EcoRl sites of the vector (Burton, D.R.
  • plgGl the heavy and light chain gene fragments were excised from the phagemids and inserted sequentially into the XhollBstEll and SacVXbal sites of the vector.
  • the recombinant plasmids were transfected into Chinese hamster ovarian (CHO) cells. Stable cell clones were established by selection with L-methionine sulfoxide (MSX) and by limiting dilution. Cell clones expressing high IgG levels were amplified and the IgGs were purified using a protein A-agarose column (Pharmacia).
  • GST-El E2 (8 ⁇ g/mL) captured by pre-coated goat anti-GST-antibody (10 ⁇ g/mL), or with ovalbumin (4 ⁇ g/mL). Specific binding was detected by alkaline phosphatase (AP)-conjugated goat anti-human IgG F(ab') 2 antibody (Pierce) (1 :500) in 1% BSA/PBS and disodium /j-nitrophenyl phosphate (Sigma), (ii) To study the relationship of different ARs to the mouse MAb epitope H53 (Cocquerel et al., J. Virol.
  • Non-fat milk 4%, BioRad
  • PBS lectin-captured antigens
  • the ELISA plates were washed after a 1 hour incubation and binding of human Fabs was detected by peroxidase (HRP)-conjugated goat anti-human IgG F(ab') 2 antibody (1 :2000) (Pierce) and TMB substrate (Pierce).
  • HRP peroxidase
  • TMB substrate TMB substrate
  • vaccinia-expressed E1E2 was either captured directly onto ELISA wells pre- coated with lectin (folded protein), or unfolded with 0.1% SDS, 50 mM DTT and incubated at 100 0 C for 5 minutes before capture onto ELISA wells (unfolded protein). Binding of the MAbs to folded and unfolded proteins was detected using the peroxidase system.
  • Mouse MAb A4 Dubuisson, J. et al., J. Virol.
  • CD81-LEL Two forms of recombinant CD81-LEL, either in fusion with glutathione S-transferase (GST) (Owsianka, A.M. et al, J. Virol. 80, 8695- 8704 (2006)) or maltose binding protein (MBP) (Chan-Fook, C.
  • GST glutathione S-transferase
  • MBP maltose binding protein
  • the binding of human MAbs was detected by HRP-conjugated goat anti- human IgG F(ab') 2 antibody as above. Non-infected/non-transfected cell lysate were used as negative controls to determine background for each MAb.
  • MAbs recognizing linear epitopes bind to both folded and unfolded proteins but the biotinylated human MAbs bind conformational epitopes on folded E2. Consequently, competition is performed with the MAbs to linear epitopes as blocking MAbs to eliminate potential non-specific signals caused by misfolded proteins in the system. After incubation for 1 h, the ELISA plates were washed and binding of biotinylated MAbs was detected with HRP-conjugated streptavidin (1 :2000, Sigma- Aldrich) in PBS with 1% BSA and TMB substrate (Pierce).
  • HCVpp human immunodeficiency virus pp neutralization
  • HCVpp was generated by co-transfection of 293T cells with pNL4-3.1ucR-E- (Connor et al, Virology 206, 935-944 (1995); He, J. et al., J. Virol. 69, 6705-671 1 (1995)) and the corresponding expression plasmids encoding the El E2 genes at 4:1 ratio by polyethylenimine (Boussif, O. et al, Proc. Natl. Acad. Sci. U.S.A.
  • RNA in the HCV GT 1 a-infected human serum KP 140 ⁇ l was purified using a QIAamp Viral RNA Mini Kit (Qiagen).
  • First strand cDNA was generated using either a reverse primer specific to HCVIa (HCVlaOuterR, GGGATGCTGCATTGAGTA, (SEQ ID NO: 697); Lavillette, D. et al., Hepatology 41, 265-274 (2005)) or random hexamer using the Superscript III reverse transcriptase (Invitrogen).
  • the GTIa E1E2 genes were amplified by a nested PCR as described previously (Lavillette, D.
  • E1E2 proteins were confirmed by the presence of folded E2 proteins in cell lysates, prepared from 293T cells transfected with the corresponding DNA plasmids, by ELISA using MAb AR3A. Antibody protection studies. Human liver-chimeric mice were prepared as described previously. Mercer,
  • mice Only mice with serum levels of hAAT greater than 60 ⁇ g/mL at 6 weeks and 100 ⁇ g/mL at 8 weeks, an indication for successful transplantation, were used in the protection study ( ⁇ 50% of transplanted mice).
  • Mice with low level of human liver chimerism were used in preliminary experiments to measure the toxicity and kinetics of MAbs in Alb-uPA/SCID mice, and the level of human IgG present in mice injected with a genotype 1 a HCV-infected human serum KP. This serum, serially diluted from 1 : 50 to 1 :4050, did not neutralize HCVpp-H77 (data not shown).
  • HCV RNA in mouse serum was quantified by a real-time TaqMan PCR assay.
  • the two primers in the real-time PCR system were designed to produce a 194 bp PCR fragment corresponding to the 5 ' non-coding region with maximum specificity to all HCV genotypes.
  • AGGTTTAGGATTCGTGCTCAT (SEQ ID NO: 699) were designed with the aid of software Primer Express (PE biosystems) and were purchased from PE Applied Biosystems.
  • PE biosystems guanidinium thiocyanate
  • silico method Bax, R. et al., J. Clin. Microbiol. 28, 495-503 (1990) was used. Briefly, 30 ⁇ L of serum was mixed with 500 ⁇ l GuSCN lysis buffer and 20 ⁇ L size- fractionated silica particles for 15 minutes. The silica particles were pelleted and washed twice with 500 ⁇ L washing buffer, twice with 70% ethanol and once with acetone.
  • Superscript Il First-Strand Synthesis Kit (Invitrogen) was used to synthesize first- strand cDNA for PCR.
  • a 50 ⁇ L mixture contained 9 ⁇ L of template HCV cDNA, 1 x TaqMan
  • a serial dilution of HCV cDNA including 1.5 X lO 6 , 1.5 x 10 s , 1.5 x 10 4 , 1.5 x 10 3 , 1.5 x 10 2 , 1.5 x 10 1 , 1.5 x 10° IU, was used to generate a standard curve for calculation of HCV RNA copy number.
  • the dynamic range of HCV RNA detection for the two step RT-PCR procedure is 6.0 x 10 2 IU/ml to 3.0 x 10 8 IU/mL.
  • Each assay run incorporates in duplicate a negative control and an HCV RNA positive control.
  • the positive control is the OptiQual HCV RNA 1 Control purchased from AcroMetrix which has been calibrated to the WHO first International Standard for HCV RNA.
  • Statistical analysis is the OptiQual HCV RNA 1 Control purchased from AcroMetrix which has been calibrated to the WHO first International Standard for HCV RNA.
  • GraphPad Prism 4 software was used for statistical analysis of the antibody protection experiment. Animals seropositive for HCV RNA by the quantitative PCR assay at or after day 7 post-infection were scored as "infected" subjects and animals seronegative up to week 6 were scored as "censored” subjects. The scores were used to construct the Kaplan-Meier survival (infection in this case) curves to calculate statistical significance between the neutralizing antibody-treated and isotype antibody control groups by a two-tailed log rank test within the experimental period. Motulsky, H. Survival curves, in GraphPad Prism4 Statistics Guide: Statistical analyses for laboratory and clinical researchers 107-117 (GraphPad Software, San Diego, 2005).
  • a total of 115 clones that exhibit specific binding to HCV E2 glycoprotein were isolated from an antibody antigen-binding fragment (Fab) phage display library generated from a donor chronically infected with HCV (see Example 1).
  • DNA sequence analysis identified 36 distinct Fabs with 13 unique heavy chain sequences. The sequences of the 36 distinct Fabs belonging to 13 groups based on the heavy chain sequences are also shown in Table 7 below. Fabs with the same designation and * or ** have the same heavy chain but distinct light chains, e.g. Hl, Hl* and Hl** have the same heavy chain, but 3 different light chains.
  • the numbers in parenthesis denote the percentage of clones recognizing each AR in the phage-display panning. It is important to note that highly isolate-specific antibodies, e.g. those against HVRl, would unlikely be selected in this study due to the use of heterologous antigens in the panning. Fab K was excluded in this table due to its poor signal in FIG. 1.
  • ⁇ 1-E2 produced by transfected 293T cells dApparent affinity is defined as the antibody concentration required to achieve half-maximal binding in an ELISA Data shown are the means of at least two independent experiments All mAbs bind natively folded, but not reduced and denatured, E2 GTIa indicates genotype Ia, GT2a indicates genotype 2a and dashes indicate that no significant inhibition or binding was observed with the highest mAb concentration tested
  • mAbs at 50, 25, 10, 5 or 1 ⁇ g/mL were tested for virus neutralization, and the lowest antibody concentrations that reduced >50% of virus infectivity are shown Dashes indicate no or ⁇ 50% virus neutralization with 50 ⁇ g/mL mAb. Data shown are the means of at least two experiments.
  • b Neutralization of HC Vpp was determined by the reduction in luciferase activity in Huh-7 cells infected with HCVpp displaying Env from different HCV isolates.
  • the panel of HCVpps shown includes HCV Env proteins that produce a signal at least tenfold higher than the background signal induced by the control pseudotype virus generated without HCV Env cDNA
  • HCV Env proteins including CH35 (genotype Ib), UKN3A1.28c (genotype 3a), UKN6.5.8 (genotype 6) and 13 different
  • KP Env clones did not produce a consistent signal tenfold higher than background and were excluded from this analysis
  • Numbers indicate percentage of residual binding signals of biotinylated human mAbs in the presence of blocking mAbs. Origin: h, human; m, mouse; r, rat.
  • the panel of variants includes substitutions at conserved residues in the putative CD81 -binding regions of E2. Substitutions important for CD81 binding are shaded and include L413A, W420A, H421A, I422A, N423A, S424A, G523A, T526A, Y527A, W529A, G530A, D535A, V538A, N540A and F550A. (Owsianka, A.M. et al. J. Virol 80, 8695-8704 (2006)).
  • the antibody competition study shows that mAbs AP33 and 3/11 (*) recognize epitopes partially dependent on proper protein folding (Tarr, A.W. et al, Hepatology 43, 592-601 (2006)).
  • the results confirm the broad designation of the antigenic regions and suggest that the discontinuous epitopes in AR3 are formed by at least three segments between amino acids 396-424, 436—447 and 523-540; the first and third segments also contribute to the CD81 -binding domain of E2 (Owsianka, A.M. et al, J. Virol.
  • a key question is whether broadly neutralizing AR3-specific antibodies can protect against infection by heterologous HCV quasispecies.
  • the human liver-chimeric Alb-uPA/SCID mouse model was used (Kneteman, N.M. et al, Hepatology 43, 1346-1353 (2006); Lindenbach, B.D. et al, Proc. Natl. Acad. Sd. USA 103, 3805-3809 (2006)).
  • this animal model is not suitable for studying virus pathogenesis, owing to its lack of a functional adaptive immune system, the question of whether antibodies can protect against HCV challenge is appropriate.
  • Previous passive antibody studies in animal models have reported relatively high antibody concentrations are needed for protection.
  • the kinetics and tolerability were first established in the animal model for the antibodies AR3 A, AR3B and a human isotype control IgGl to HIV-I , b6.
  • Transplanted Alb-uPA/SCID mice with a low level of human liver chimerism were injected intraperitoneally with 100, 150 or 200 mg/kg MAb, and blood samples were collected by tail bleed.
  • Human antibody in the murine sera was measured by a quantitative sandwich ELISA using conjugated and unconjugated goat anti-human F(ab)' 2 antibody. The antibodies did not show adverse effects in control mice. No specific weight loss or signs of illness associated with the administration of the MAbs were noted in the mice during the experiment.
  • mice One mouse (N457) was euthanized due to unrelated morbidity at Day 7. A dose of 200 mg/kg given through intraperitoneal injection was required to achieve mean serum titers approximately 100 x higher than in vitro neutralization titers. Such titers have previously been found to be necessary to achieve sterilizing immunity in other viral disease models.
  • the neutralizing activity in mouse sera collected ten days after injection was determined by HCVpp-H77 neutralization assay.
  • Mouse sera containing anti-HCV MAbs AR3A and AR3B neutralized 50% of HCVpp infectivity (IC 5 o) in the range of 1:200 to 1:1000.
  • the IC 50 titers of mouse sera containing anti-HCV MAbs AR3A and AR3B were in the range of 0.4-1.1 (mean 0.8 ⁇ s.d. 0.3) and 0.5-3 (mean 1.2 ⁇ s.d. 0.9) ⁇ g/mL, respectively.
  • Isotype control MAbs b6 & DEN3 did not neutralize HCVpp.
  • the observed half-lives of mAbs AR3 A, AR3B and b6 were 6.0 ⁇ 2.2 d, 9.0 ⁇ 1.3 d and 7.3 ⁇ 1.8 d (mean ⁇ s.d.), respectively, and their specific neutralizing activities (that is, neutralizing activity relative to serum mAb concentration) were stable for at least 10 days in the mice.
  • the mAbs were administered intraperitoneally in passive transfer experiments to mice with high levels of human liver chimerism (see Example 1), and the mean serum titers of mAbs AR3A, AR3B and the control mAb b6, at 24 hours after injection were -2.5 ⁇ 0.3 mg/mL, 3.1 ⁇ 0.5 mg/mL and 2.6 ⁇ 0.3 mg/mL, respectively (FIG. 4).
  • the partial amino acid sequences (residues 384-622) of forty HCVs found in the viral quasispecies population in the HCV genotype la-infected human serum are shown below.
  • HCV E2 glycoprotein is a major target for virus neutralizing antibodies and an important component in a HCV vaccine.
  • E2 has encoded several features to evade antibodies. First, E2 encodes regions that are highly mutable. Rapid changes in viral sequence facilitate virus escape. Second, E2 is highly glycosylated and the associated glycans help shield the neutralizing epitopes from antibodies. Despite these escape features, we have identified the antigenic region 3 (AR3) on E2 as a relatively conserved target for antibody neutralization in vitro and antibody protection in vivo. The amino acid residues important for the binding of AR3-specific antibodies is described above. The following show how these residues organize together to form the AR3 conformational epitopes.
  • AR3 antigenic region 3
  • E2 that displays AR3 properly while silencing some of the variable sequences that are usually immunogenic but are not targets of broadly neutralizing antibodies
  • a panel of E2 truncation mutants were constructed.
  • the minimal E2 fragment that displays the CD81 -binding sites and the broadly neutralizing epitopes correctly the binding of these E2 mutants with CDE81- LEL or various mAb were studied.
  • E2 mutants were constructed by deletion of highly variable regions, specific N-glycosylation signals, or every other cysteine residues from C- or N- terminus of wildtype (WT) E2.
  • WT wildtype
  • the panel of E2 mutants in fusion with the Flag tags at their C-termini are illustrated in FIG. 7, and their sequences are shown in Table 16.
  • Table 16 Hepatitis C virus E2 Glycoprotein Mutants
  • the cDNA encoding these mutants were generated by polymerase chain reaction (PCR) or by splicing by overlap extension polymerase chain reaction (SOE- PCR) as described in Horton et al., Biotechniques 8:528-535 (1990).
  • PCR polymerase chain reaction
  • SOE- PCR overlap extension polymerase chain reaction
  • the plasmid pCV-H77c (Genbank accession# AFOl 1751) encoding wildtype E2 gene of the isolate H77 was used as a template.
  • the primers used in the reactions are enlisted below.
  • E2wtF 744 AATAACGCGTGAAACCCACGTCACCGG
  • E2flF 745 AATAACGCGTCAACTGATCAACACCAACG
  • E2f2F 746 AATAACGCGTTTGGCCAGCTGCCGACGC
  • E2f4F 748 AATAACGCGTGTATATTGCTTCACTCCCAG
  • E2f5F 749 AATAACGCGTACTGGATTCACCAAAGTGTG
  • E2wtR 750 TATTCTCGAGCTCCCACTTAATGGCCCAG
  • E2r2 752 TATTCTCGAGCCGCGTCCAGTTGCAGGC
  • E2r2a 753 TATTCTCGAGGTTGCAGGCCGCTTCCAGC
  • E2r3 754 TATTCTCGAGGTAGTCGACCATGCACCTG
  • E2r5 756 TATTCTCGAGCACCCCTCCGATGACACAAG
  • E2r7 758 TATTCTCGAGCGGGCCACACACGCTCTTTG delHVR2F 759 TGCGGCTCTAGCGGATGCTGGCACTACCCTCCAAG delHVR2R 760 CAGCATCCGCTAGAGCCGCAGCTGGCCAACCTCTC delHVR3F 761 TGTGGAAGCTCTGGCTGCCCCACTGATTGCTTCC delHVR3R 762 GCAGCCAGAGCTTCCACAAGGGGGCGCTCCGCAC
  • PCR conditions 94°C, 3 min; 25 cycles of (94°C, 30 s; 55°C, 30 s; 70 0 C, 90 s); & 70 0 C, 10 min; PCR system: Platinum Pfx DNA polymerase (Invitrogen); PCR instrument: GeneAmp PCR System 9700 (Applied Biosystems).
  • the PCR products generated in Table 18 were resolved by agarose gel electroporesis and the DNA bands of correct size were excised and purified.
  • the products were either used as templates in a second PCR, or were digested with MIu I and Xho I restriction enzymes.
  • the digested products were gel-purified and inserted between the BssH II and Xho I sites of the plasmid pCMV-Tag4A-tpaJR-FLgpl20 (Pantophlet et al., J Virol 77:642-658 (2003); Law et ai, J Virol 81 :4272-4285 (2007)).
  • the inserted products replaced the HIV genes in the plasmid and are in frame with a 5 '-signal peptide and a 3'-FLAG tag to facilitate protein secretion and for detection.
  • the nucleotide sequences of the E2 mutants were verified by DNA sequencing.
  • E2 mutants were expressed by transient transfection of 293T cells.
  • Cell monolayers were co-transfected with the expression plasmids encoding the different E2 mutants and pAdVAntage plasmid (Promega) at 1:1 ratio by polyethylenimine (Boxxssif et al., Proc Natl Acad Sd USA 92:7297-7301 (1995)).
  • Cell supernatants were collected 3 days post-transfection and were clarified by centrifugation.
  • MAbs monoclonal antibodies
  • HCV co-receptor CD81 HCV co-receptor CD81
  • Mutants with correctly folded antibody epitopes or CD81 -binding sites were captured by the corresponding reagents and the captured mutants were detected with a mouse anti-FLAG tag MAb (Sigma), followed by a secondary antibody (Peroxidase- conjugated AffiniPure Goat Anti-mouse IgG from Jackson ImmunoResearch Laboratories) and the colorimetic peroxidase substrate TMB (Pierce). The peroxidase reaction was stopped by adding sulfuric acid.
  • E2 mutants Specific binding of the E2 mutants to the capturing reagents were detected by measuring the absorbances of the samples at 450 nm using a microplate reader (Molecular Devices). The results are summarized in FIG.8.
  • the CD81 -binding sites and AR3 are presented well on the E2 mutants E2 ⁇ TM, E2flrl, E2flr2, E2flr2a, E2 ⁇ N5 and E2 ⁇ N9.
  • the mutant E2 ⁇ N5N9 was captured by MAbs AR3A or AR3C at a comparable level to the above mutants but at a much reduced level by CD81-LEL, MAbs AR3B or AR3D.
  • mutants E2flr3, E2flr4, E2flr5, E2f2r2a and E2f3r2a were captured by the non-neutralizing MAbs ARIA and ARlB but not CD81-LEL or AR3-specific MAbs, suggesting that the CD81 -binding sites and the broadly neutralizing epitopes in AR3 are not present or folded correctly in these mutants.
  • fragments E2flrl and E2flr2abind to the conformation-dependent, broadly neutralizing MAb AR3A and CD81-LEL indicates that the E2 residues 412-645 and cysteines 1-16 are important for correct folding of AR3 (within this region, residues 460-485 and 570-580 are not required).
  • E2 ⁇ TM binds all Abs recognizing ARl , 2 and 3, but weakly to CD81-LEL.
  • the HCV envelope El and E2 glycoproteins are technically challenging to produce as El does not fold properly in the absence of E2 (Michalak et al, J Gen Virol 78:2299-2306 (1997) and Patel et al, Virology 279:58-68 (2001)) and efficient production of E2 is influenced by El (Cocquerel et al., J Virol 77:10677- 10683 (2003), Brazzoli et al, Virology 332:438-453 (2005)).
  • E2661 A truncated version of E2 (known as E2661) can be expressed independently and retain its function in binding to the co-receptor CD81 (Michalak et al., J Gen Virol 78:2299-2306 (1997); Flint et al., J Virol 73 :6235-6244 (1999); Flint et al, J Virol 74:702-709 (2000)).
  • This truncated E2 has not been shown to be produced in a highly purified form suitable for biochemical analysis and crystallization attempts (Flint et al., J Virol 74:702-709 (2000)).
  • E2 displaying correctly folded AR3 epitopes a protein production and purification method was developed.
  • the plasmids encoding the E2 mutants pE2 ⁇ TM and pE2flr2a were co-transfected with pAdVAntage plasmid (Promega) at 1:1 ratio into FreeStyle 293 cells (Invitrogen) using 293fectin Transfection Reagent (Invitrogen). Cell supernatants were collected twice at 3-day and 5-day post- transfection.
  • kifunensine (at 7.5 ⁇ M, Cayman Chemical) (Elbein et al., J Biol Chem 265:15599-15605 (1990); Chang et al., Structure 15:267-273 (2007)) was added to cell culture media to improve glycan homogeneity on E2.
  • the E2 mutants were purified by antibody affinity chromatography.
  • the MAb AR3A which can distinguish folded from misfolded protein, was used.
  • the MAb AR3A recognizes a conformation-dependent epitope on E2, neutralizes HCV in vitro and offers protection against HCV infection in vivo as shown above.
  • MAb AR3A binds natively folded E2 at high affinity but not denatured and reduced E2.
  • MAb AR3A-affinity matrix MAb AR3A was first captured by Protein A-Sepharose (GE Healthcare) at a ratio of 10 mg MAb per mL Sepharose beads. After overnight incubation, the beads were washed 3 times with 0.2 M sodium borate buffer (pH 9). MAb AR3A was then crosslinked chemically to the Protein A-beads using dimethyl pimelimidate (Thermo Scientific). The reaction was stopped after lhour incubation at room temperature by pelleting the beads and washing the beads 3 times with 0.2 M ethanolamine (pH 8).
  • the MAb-conjugated beads were packed into an Econo-Column (Bio-Rad) and the beads were rinsed once with 0.2 M glycine (pH 2.2) followed by PBS to equilibrate the column for affinity purification of the E2 mutants.
  • Cell supernatants containing the E2 mutants were clarified by low-speed centrifugation and filtration through a 0.22- ⁇ m filter before loading onto the affinity columns by gravity flow. The flow-through solutions were collected and the columns were washed with PBS. Bound proteins were released from the affinity columns using different elution conditions and the antigenicity of the eluted proteins were investigated (see below).
  • the eluants were concentrated and monomers of the E2 mutants were purified by size-exclusion chromatography using a Superdex 75 column (Amersham Biosciences).
  • E2flr2a Three batches of E2flr2a were produced by transient transfection of 293T cells ( ⁇ 5 x 10 8 cells per batch) with the corresponding expression plasmid.
  • Batch 3 was produced in the presence of 10 ⁇ M kifunesine (BIOMOL), a potent inhibitor of the glycoprotein processing ⁇ -mannosidase I and is used to improve glycan homogeneity in the glycoproteins.
  • Cell supernatants were loaded onto an antibody- affinity column (MAb AR3A, 5 mL) by gravity flow and bound proteins were eluted with a low pH buffer (0.2 M glycine, pH 2.7). Batches 2 and 3 was purified twice to monitor purification efficiency.
  • E2flr2a was further purified and analyzed by size-exclusion chromatography.
  • E2flr2a purified by MAb AR3A affinity column was concentrated to 0.5 mL using an ultra-centrifugal filter device with a 30 kDa nominal molecular weight limit (Millipore).
  • the concentrated proteins were loaded onto a Sephadex 75 size- exclusion column (GE Healthcare) using a AKTA Fast Protein Liquid Chromatography (FPLC) system (GE Healthcare).
  • the proteins were separated in Tris buffer (0.1 M Tris-HCl pH 7.4 and 150 mM NaCl) and elution fractions of 0.5 mL were collected by an automatic fractionator.
  • the chromatogram of E2flr2a was compared to the chromatogram of protein standards, including: (A) blue dextran 2000, (B) bovine serum albumin 67 kDa, (C) ovalbumin 43 kDa, and (D) chymotrypsinogen 25 kDa (GE Healthcare). Fractions 14-22 were analyzed by non-reducing SDS- PAGE (4-15% gradient, BIO-RAD). The results showed that the high molecular weight impurities eluted from the MAb AR3A-afflnity column were separated from the glycoforms of E2flr2a, which appear to be monomers of size between 43-67 kDa in gel filtration.
  • Reaction conditions included (1) E2flr2a eluted with low pH buffer as a control; (2) E2flr2a eluted with 0.5 M NaSCN; (3) 1 M NaSCN; and (4) 2 M NaSCN.
  • the purified proteins were analyzed by non-reducing SDS- PAGE (4-15% gradient, BIO-RAD). The results show that neutral pH elution conditions can be used to purify the E2flr2a glycoforms, with the exception of elution with 0.5 M NaSCN, which led to high molecular weight impurities.
  • E2flr2a was produced in the presence of kifunensine, loaded onto a MAb AR3A-affinity column and eluted with a step-gradient of buffers with increasing pH.
  • the eluents were collected into tubes with 0.1 volume of neutralizing buffer (2 M T ⁇ s-HCl, pH 7.4).
  • Reaction conditions included (1) E2flr2a eluted with 2 M NaSCN as a control; (2) E2flr2a eluted with 0.2 M glycine pH 9.5; (3) pH 10.5; (4) pH 11.5; (5) pH 12.5; and (6) pH 11.5 sample filtered through an ultra-centrifugal filter device with a 100 kDa nominal molecular weight limit (Milhpore).
  • the results show that both 2 M NaSCN and 0 2 M glycine pH 11.5 elution conditions led to pure E2flr2a protein.
  • E2 ⁇ TM was purified using a MAb AR3A-conjugated affinity column with a 2M NaSCN, pH 7.4 elution and analyzed by SDS-PAGE.
  • High molecular weight impurities were removed by filtering through an ultracentrifugal filter device with a 100 kDa nominal molecular weight limit (Milhpore).
  • the purified proteins were analyzed by 4-15% gradient non-reducing SDS-PAGE (BIO-RAD). The results show that ultracentrifugal filtration removed high molecular weight impurities in samples produced in both the absence and presence of kifunensine. Protein concentration was quantified by the Bradford method (Bradford et al.,
  • the recombinant E2 fragment E2flr2a can be purified to greater than 90 % by a single affinity chromatography step.
  • the purification method is applicable to E2flr2a produced in the presence of the plant alkaloid kifunensine, a potent inhibitor of the glycoprotein processing ⁇ -mannosidase I.
  • N-glycans on recombinant proteins produced in the presence of kifunensine are almost exclusively high-mannose type oligosaccharides, which can be readily trimmed by endoglycosidase H digestion to improve protein homogeneity.
  • E2 mutants E2 ⁇ TM and E2flr2a can be purified as monomers.
  • the recombinant E2 fragments purified by the above method adopt a native fold as found on viral surface.
  • the purified E2 mutants will be extremely useful in research and discovery of anti-viral drugs and HCV vaccines.

Abstract

La présente invention concerne des polypeptides E2 mutants utilisés à des fins thérapeutiques et diagnostiques. Leur séquence d'acides aminés comprend, de la terminaison amino à la terminaison carboxy: (1) un premier segment qui correspond aux résidus d'acides aminés 412 à 459 du polypeptide E2 d'un virus de l'hépatite C spécifique, (2) un deuxième segment qui correspond aux résidus d'acides aminés 486 à 569 du polypeptide E2 du virus de l'hépatite C spécifique, et (3) un troisième segment qui correspond aux résidus d'acides aminés 581 à 645 du polypeptide E2 du virus de l'hépatite C spécifique, les segments étant liés directement ou via un lieur de polypeptide, à condition que le polypeptide mutant ne comporte pas de séquence contiguë d'acides aminés correspondant à tous les résidus d'acides aminés 411 à 662 du polypeptide E2 du virus de l'hépatite C spécifique; le polypeptide mutant comporte des suppressions de résidus d'acides aminés correspondant aux acides aminés 384 à 411 et 718 à 746 du polypeptide E2 pleine longueur du virus de l'hépatite C spécifique et le polypeptide mutant peut se lier spécifiquement à un anticorps qui se lie avec un épitope conformationnel sur le polypeptide E2 contenant au moins les acides aminés 411 à 462. Le polypeptide E2 mutant susmentionné peut être utilisé pour la production d'anticorps à neutralisation croisée contre le virus de l'hépatite C. La présente invention concerne également des préparations et des compositions pharmaceutiques contenant des polypeptides E2 mutants et des procédés permettant leur préparation, leur purification et leur utilisation. L'invention concerne également des procédés consistant à utiliser les polypeptides E2 mutants pour prévenir et traiter l'infection par le virus de l'hépatite C.
PCT/US2009/005785 2008-10-24 2009-10-23 Polypeptides e2 mutants du virus de l'hépatite c pour le traitement du virus de l'hépatite c WO2010047829A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19727308P 2008-10-24 2008-10-24
US61/197,273 2008-10-24

Publications (1)

Publication Number Publication Date
WO2010047829A1 true WO2010047829A1 (fr) 2010-04-29

Family

ID=41531756

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/005785 WO2010047829A1 (fr) 2008-10-24 2009-10-23 Polypeptides e2 mutants du virus de l'hépatite c pour le traitement du virus de l'hépatite c

Country Status (1)

Country Link
WO (1) WO2010047829A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016036252A3 (fr) * 2014-09-05 2016-05-19 Aimm Therapeutics B.V. Anticorps spécifique du virus de l'hépatite c
EP2868665A4 (fr) * 2013-02-27 2016-06-01 Univ Peking Peoples Hospital Peptide épitopique puhi26 de lymphocytes b dirigé contre le virus de l'hépatite c et ses applications
US20170121390A1 (en) * 2013-12-27 2017-05-04 Tatiana Nikolaevna Vlasik Method for neutralizing hepatitis c virus, fully human monoclonal antibody against hepatitis c virus (variants), composition of fully human monoclonal antibodies against hepatitis c virus and hybrid mouse/human producer cell line of fully human monoclonal antibodies against hepatitis c virus (variants)
WO2021021603A3 (fr) * 2019-07-26 2021-04-22 The Scripps Research Institute Immunogènes e2 de vhc modifiés et compositions de vaccin associées

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3561444A (en) 1968-05-22 1971-02-09 Bio Logics Inc Ultrasonic drug nebulizer
US3703173A (en) 1970-12-31 1972-11-21 Ted A Dixon Nebulizer and tent assembly
US4624251A (en) 1984-09-13 1986-11-25 Riker Laboratories, Inc. Apparatus for administering a nebulized substance
US4635627A (en) 1984-09-13 1987-01-13 Riker Laboratories, Inc. Apparatus and method
EP1178116A1 (fr) * 2000-08-03 2002-02-06 Hybrigenics S.A. Acides nucléiques Sid et polypeptides sélectionnés à partir d'une souche pathogène du virus de l' hépatite ainsi que leurs applications
WO2004005316A2 (fr) * 2002-07-02 2004-01-15 Board Of Trustees Of Leland Stanford Jr. University Prevention et traitement d'une infection de vch faisant appel a des anticorps diriges contre des epitopes conformationnels et lineaires
WO2005012502A2 (fr) * 2003-03-28 2005-02-10 Idm Pharma, Inc. Procedes d'identification de variants optimaux d'epitopes peptidiques
WO2009061739A1 (fr) * 2007-11-06 2009-05-14 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Neutralisation du vhc

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3561444A (en) 1968-05-22 1971-02-09 Bio Logics Inc Ultrasonic drug nebulizer
US3703173A (en) 1970-12-31 1972-11-21 Ted A Dixon Nebulizer and tent assembly
US4624251A (en) 1984-09-13 1986-11-25 Riker Laboratories, Inc. Apparatus for administering a nebulized substance
US4635627A (en) 1984-09-13 1987-01-13 Riker Laboratories, Inc. Apparatus and method
EP1178116A1 (fr) * 2000-08-03 2002-02-06 Hybrigenics S.A. Acides nucléiques Sid et polypeptides sélectionnés à partir d'une souche pathogène du virus de l' hépatite ainsi que leurs applications
WO2004005316A2 (fr) * 2002-07-02 2004-01-15 Board Of Trustees Of Leland Stanford Jr. University Prevention et traitement d'une infection de vch faisant appel a des anticorps diriges contre des epitopes conformationnels et lineaires
WO2005012502A2 (fr) * 2003-03-28 2005-02-10 Idm Pharma, Inc. Procedes d'identification de variants optimaux d'epitopes peptidiques
WO2009061739A1 (fr) * 2007-11-06 2009-05-14 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Neutralisation du vhc

Non-Patent Citations (47)

* Cited by examiner, † Cited by third party
Title
A. TAKAMIZAWA ET AL., J VIROL., vol. 65, 1991, pages 1105 - 1113
ABU DAYYEH BK, CHUNG RT.: "Broadly neutralizing antibodies against hepatitis C virus: new hope for immunization?", GASTROENTEROLOGY,ELSEVIER, PHILADELPHIA, PA, vol. 134, no. 7, 13 May 2008 (2008-05-13), pages 2184 - 2186, XP022756870 *
BIOCHEM., vol. 11, no. 9, 1972, pages 1726 - 1732
BURIONI R ET AL: "Cross-reactive pseudovirus-neutralizing anti-envelope antibodies coexist with antibodies devoid of such activity in persistent hepatitis C virus infection", VIROLOGY, ACADEMIC PRESS,ORLANDO, US, vol. 327, no. 2, 1 October 2004 (2004-10-01), pages 242 - 248, XP004537624, ISSN: 0042-6822 *
BURIONI R, PEROTTI M, MANCINI N, CLEMENTI M.: "Perspectives for the utilization of neutralizing human monoclonal antibodies as anti-HCV drugs.", J HEPATOL., vol. 49, no. 2, August 2008 (2008-08-01), pages 299 - 302, XP002565590 *
CARILLO ET AL., APPLIED MATH., vol. 48, 1988, pages 1073
CHAN-FOOK ET AL., VIROLOGY, vol. 273, 2000, pages 60 - 66
CLAYTON, R.F. ET AL., J VIROL., vol. 76, 2002, pages 7672 - 7682
DELEERSNYDER, V. ET AL., J VIROL., vol. 71, 1997, pages 697 - 704
DUBUISSON, J. ET AL., J VIROL., vol. 68, 1994, pages 6147 - 6160
EREN R, ET AL: "Preclinical evaluation of two neutralizing human monoclonal antibodies against hepatitis C virus (HCV): a potential treatment to prevent HCV reinfection in liver transplant patients.", J VIROL., vol. 80, no. 6, March 2006 (2006-03-01), pages 2654 - 2664, XP009068539 *
FLINT, M. ET AL., J VIROL., vol. 73, 1999, pages 6235 - 6244
GAUDUIN ET AL., NAT. MED., vol. 3, 1997, pages 1389 - 1393
GRIBSKOV, M. AND DEVEREUX, J.,: "Sequence Analysis Primer", 1991, M. STOCKTON PRESS
GRIFFIN, A.M., AND GRIFFIN, H. G.,: "Computer Analysis of Sequence Data, Part I", 1994, HUMANA PRESS
HSU, M. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 100, 2003, pages 7271 - 7276
KECK Z Y ET AL: "Therapeutic control of hepatitis C virus: the role of neutralizing monoclonal antibodies", CURRENT TOPICS IN MICROBIOLOGY AND IMMUNOLOGY, SPRINGER, BERLIN, DE, vol. 317, 1 January 2008 (2008-01-01), pages 1 - 38, XP009102540, ISSN: 0070-217X *
KECK ZY, LI SH, XIA J, VON HAHN T, BALFE P, MCKEATING JA, WITTEVELDT J, PATEL AH, ALTER H, RICE CM, FOUNG SK.: "Mutations in hepatitis C virus E2 located outside the CD81 binding sites lead to escape from broadly neutralizing antibodies but compromise virus infectivity.", VIROL., vol. 83, no. 12, 2009, XP002565592 *
KECK, Z.Y. ET AL., J VIROL., vol. 78, 2004, pages 9224 - 9232
KECK, Z.Y. ET AL., J. VIROL., vol. 79, 2005, pages 13199 - 13208
KECK, Z.Y. ET AL., J. VIROL., vol. 81, 2007, pages 1043 - 1047
KNETEMAN, N.M. ET AL., HEPATOLOGY, vol. 43, 2006, pages 1346 - 1353
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 - 97
KOZBOR ET AL., IMMUNOL TODAY, vol. 4, 1983, pages 72
LAW M ET AL.: "Broadly neutralizing antibodies protect against hepatitis C virus quasispecies challenge.", NAT MED., vol. 14, no. 1, 6 December 2007 (2007-12-06) - January 2008 (2008-01-01), pages 25 - 27, XP002563296 *
LESK, A. N.,: "Computational Molecular Biology", 1988, OXFORD UNIVERSITY PRESS
LINDENBACH, B.D. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 3805 - 3809
MARUYAMA, T. ET AL., AM. J PATHOL., vol. 165, 2004, pages 53 - 61
MARUYAMA, T. ET AL., AM. J. PATHOL., vol. 165, 2004, pages 53 - 61
MEYER K, AIT-GOUGHOULTE M, KECK ZY, FOUNG S, RAY R.: "Antibody-dependent enhancement of hepatitis C virus infection.", J VIROL., vol. 82, no. 5, 19 December 2007 (2007-12-19) - March 2008 (2008-03-01), pages 2140 - 2149, XP002565591 *
MORRISON ET AL., PROC. NAT. ACAD. SCI. US.A., vol. 86, pages 6851
N. KATO ET AL., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 9524 - 9528
OWSIANKA, A. ET AL., J VIROL., vol. 79, 2005, pages 11095 - 11104
OWSIANKA, A.M. ET AL., J VIROL., vol. 80, 2006, pages 8695 - 8704
PARREN ET AL., J VIROL., vol. 75, 2001, pages 8340 - 8347
Q.-L. CHOO ET AL., PROC. NATL. ACAD. SCI. USA, vol. 88, 1991, pages 2451 - 2455
SLATER-HANDSHY T, DROLL DA, FAN X, DI BISCEGLIE AM, CHAMBERS TJ.: "HCV E2 glycoprotein: mutagenesis of N-linked glycosylation sites and its effects on E2 expression and processing.", VIROLOGY., vol. 319, no. 1, 5 February 2004 (2004-02-05), pages 36 - 48, XP002565589 *
SMITH, D.W.,: "Biocomputing: Informatics and Genome Projects", 1993, ACADEMIC PRESS
STAMATAKI Z, ET AL.: "Hepatitis C virus envelope glycoprotein immunization of rodents elicits cross-reactive neutralizing antibodies.", VACCINE, vol. 25, no. 45, 7 November 2007 (2007-11-07), pages 7773 - 7784, XP002565588 *
TARR ALEXANDER W ET AL: "Determination of the human antibody response to the epitope defined by the hepatitis C virusneutralizing monoclonal antibody AP33", JOURNAL OF GENERAL VIROLOGY, SOCIETY FOR GENERAL MICROBIOLOGY, SPENCERS WOOD, GB, vol. 88, no. Part 11, 1 November 2007 (2007-11-01), pages 2991 - 3001, XP002515099, ISSN: 0022-1317 *
TARR, A.W. ET AL., HEPATOLOGY, vol. 43, 2006, pages 592 - 601
VON HEINGE, G.: "Sequence Analysis in Molecular Biology", 1987, ACADEMIC PRESS
WATSON ET AL.: "Molecular Biology of the Gene, 4th Edition,", 1987, THE BENJAMIN/CUMMINGS PUB. CO., pages: 224
WEILER ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 2792 - 2799
ZHANG PEI ET AL.: "Depletion of interfering antibodies in chronic hepatitis C patients and vaccinated chimpanzees reveals broad cross-genotype neutralizing activity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 106, no. 18, May 2009 (2009-05-01), pages 7537 - 7541, XP002565593 *
ZHANG PEI ET AL: "Hepatitis C virus epitope-specific neutralizing antibodies in Igs prepared from human plasma", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC, US, vol. 104, no. 20, 15 May 2007 (2007-05-15), pages 8449 - 8454, XP002515098, ISSN: 0027-8424 *
ZHONG, J. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 102, 2005, pages 9294 - 9299

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2868665A4 (fr) * 2013-02-27 2016-06-01 Univ Peking Peoples Hospital Peptide épitopique puhi26 de lymphocytes b dirigé contre le virus de l'hépatite c et ses applications
US20170121390A1 (en) * 2013-12-27 2017-05-04 Tatiana Nikolaevna Vlasik Method for neutralizing hepatitis c virus, fully human monoclonal antibody against hepatitis c virus (variants), composition of fully human monoclonal antibodies against hepatitis c virus and hybrid mouse/human producer cell line of fully human monoclonal antibodies against hepatitis c virus (variants)
US10633431B2 (en) * 2013-12-27 2020-04-28 Tatiana Nikolaevna Vlasik Method for neutralizing hepatitis C virus, fully human monoclonal antibody against hepatitis C virus (variants), composition of fully human monoclonal antibodies against hepatitis C virus and hybrid mouse/human producer cell line of fully human monoclonal antibodies against hepatitis C virus (variants)
WO2016036252A3 (fr) * 2014-09-05 2016-05-19 Aimm Therapeutics B.V. Anticorps spécifique du virus de l'hépatite c
US10435463B2 (en) 2014-09-05 2019-10-08 Academisch Medisch Centrum Hepatitis C virus specific antibody
WO2021021603A3 (fr) * 2019-07-26 2021-04-22 The Scripps Research Institute Immunogènes e2 de vhc modifiés et compositions de vaccin associées
US11008368B2 (en) * 2019-07-26 2021-05-18 The Scripps Research Institute Engineered HCV E2 immunogens and related vaccine compositions
CN114630698A (zh) * 2019-07-26 2022-06-14 斯克里普斯研究学院 经改造hcv e2免疫原和相关疫苗组合物
US20230034467A1 (en) * 2019-07-26 2023-02-02 The Scripps Research Institute Engineered HCV E2 Immunogens and Related Vaccine Compositions

Similar Documents

Publication Publication Date Title
Wang et al. Immunodominant SARS coronavirus epitopes in humans elicited both enhancing and neutralizing effects on infection in non-human primates
US8858947B2 (en) Hepatitis C antibodies and uses thereof
KR20180012245A (ko) 다가 b형 간염 바이러스 항원 결합 분자 및 그의 용도
AU2016250188B2 (en) Antibody-mediated neutralization of Chikungunya virus
CN106589116B (zh) 一种黄病毒人源单克隆抗体及应用
WO2000005266A1 (fr) Anticorps contre le virus de l'hepatite c et ses utilisations
TW201444866A (zh) 針對呼吸道融合病毒f蛋白質的人類抗體及其使用方法
CN107973850B (zh) 重组猪瘟病毒e2蛋白猪源化单克隆抗体及其制备方法和应用
US20110311550A1 (en) Agents for hcv treatment
CN115768790A (zh) 针对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的人单克隆抗体
Dong et al. Candidate peptide-vaccine induced potent protection against CSFV and identified a principal sequential neutralizing determinant on E2
Krapchev et al. Recombinant Flag-tagged E1E2 glycoproteins from three hepatitis C virus genotypes are biologically functional and elicit cross-reactive neutralizing antibodies in mice
US20100104555A1 (en) HCV neutralizing epitopes
EP2376119A1 (fr) Anticorps monoclonal anti-vhc en tant que médicament dans le traitement prophylactique et thérapeutique des infections par le vhc
WO2010047829A1 (fr) Polypeptides e2 mutants du virus de l'hépatite c pour le traitement du virus de l'hépatite c
CN104324373B (zh) 组合物及其制备方法
JP2004524829A (ja) C型肝炎ウイルスのe2糖タンパク質に特異的なモノクローナル抗体、ならびにc型肝炎の診断、治療、および予防におけるそれらの使用
Torresi et al. A self‐adjuvanting multiepitope immunogen that induces a broadly cross‐reactive antibody to hepatitis C virus
CN115087667B (zh) 特异性结合SARS-CoV-2的抗原结合蛋白
CN107286237B (zh) 一种抗丙型肝炎病毒抗体的获取以及应用
Liao et al. Hepatitis B virus precore protein augments genetic immunizations of the truncated hepatitis C virus core in BALB/c mice
CN114716541B (zh) 抗冠状病毒的全人广谱中和抗体76e1及其应用
CN112239498B (zh) 一种抗丙型肝炎病毒抗体
CN112239497B (zh) 一种抗丙型肝炎病毒抗体
RU2803082C2 (ru) Антитела против вируса гепатита в и их применение

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09744235

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09744235

Country of ref document: EP

Kind code of ref document: A1