WO2016141201A2 - Conception rationnelle de vaccins contre le virus de l'hépatite c - Google Patents

Conception rationnelle de vaccins contre le virus de l'hépatite c Download PDF

Info

Publication number
WO2016141201A2
WO2016141201A2 PCT/US2016/020720 US2016020720W WO2016141201A2 WO 2016141201 A2 WO2016141201 A2 WO 2016141201A2 US 2016020720 W US2016020720 W US 2016020720W WO 2016141201 A2 WO2016141201 A2 WO 2016141201A2
Authority
WO
WIPO (PCT)
Prior art keywords
hcv
polypeptide
residues
binding
virus
Prior art date
Application number
PCT/US2016/020720
Other languages
English (en)
Other versions
WO2016141201A3 (fr
Inventor
Roy A. MARIUZZA
Yili LI
Thomas R. Fuerst
Brian G. Pierce
Steven K. H. Foung
Zhen-Yong Keck
Original Assignee
University Of Maryland
The Board Of Trustees Of The Leland Stanford Junior University
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 University Of Maryland, The Board Of Trustees Of The Leland Stanford Junior University filed Critical University Of Maryland
Publication of WO2016141201A2 publication Critical patent/WO2016141201A2/fr
Publication of WO2016141201A3 publication Critical patent/WO2016141201A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1081Togaviridae, e.g. flavivirus, rubella virus, hog cholera virus
    • C07K16/109Hepatitis C virus; Hepatitis G virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • 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
    • 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
    • C12N2770/24234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • HCV Hepatitis C virus
  • a challenge for HCV vaccine development is to identify conserved epitopes able to elicit protective antibodies against this highly diverse virus.
  • Glycan shielding is a mechanism by which HCV masks such epitopes on its E2 envelope glycoprotein.
  • Antibodies to the E2 region comprising residues 412-423 (E2412-423) have broadly neutralizing activities.
  • an adaptive mutation in this linear epitope, Asn417Ser is associated with a glycosylation shift from Asn417 to Asn415 that enables HCV to escape neutralization by monoclonal antibodies (mAbs) such as HCV1 and AP33.
  • mAbs monoclonal antibodies
  • the human mAb HC33.1 can neutralize virus bearing the Asn417Ser mutation.
  • HC33.1 penetrates the glycan shield created by the glycosylation shift to Asn415
  • the conformation of E2412-423 bound to HC33.1 is distinct from the ⁇ -hairpin conformation of this peptide bound to HCV1 or AP33, due to disruption of the ⁇ -hairpin through interactions with the unusually long complementarity- determining region 3 (CDR3) of the HC33.1 heavy chain.
  • CDR3 complementarity- determining region 3
  • HCV Hepatitis C virus
  • Flaviviridae family of positive-stranded RNA viruses infects at least 2% of the world population and is a major cause of liver cirrhosis, liver failure, and hepatocellular carcinoma.
  • the global burden is estimated at 170 million infected individuals with an annual increase of 3-4 million new infections.
  • DAAs direct-acting antiviral agents
  • HCV vaccine The key challenge in HCV vaccine development is to overcome the high diversity of this virus and its potential to escape from host immune responses.
  • HCV is composed of a nucleocapsid core enveloped by a lipid bilayer in which two surface glycoproteins, E1 and E2, are anchored. Entry of HCV into hepatocytes is mediated by interactions between the E1E2 heterodimer and at least four cellular receptors: the tetraspanin CD81 , scavenger receptor class B type 1 (SR-B1), and the tight junction proteins occludin and claudin 1.
  • An effective vaccine must include conserved epitopes of E1E2 that are able to elicit broadly neutralizing antibodies.
  • E1E2 monoclonal antibodies
  • E2 contains binding sites for CD81 and SR-B1.
  • a significant challenge for vaccine development is defining conserved epitopes that i) are capable of eliciting protective antibodies in this highly diverse virus, and ii) are resistant to development of escape mutants.
  • Treatment of HCV and the development of vaccines that broadly protect against highly diverse HCV genotypes and subtypes are of interest in the field.
  • the present invention addresses this issue.
  • HCV E2 glycoprotein which is the major target of neutralizing antibody response to virus, is modified to enhance the protective immune response.
  • modified polypeptides which may be referred to as an HCV E2 antigen, or antigenic polypeptide, are typically at least about 50 amino acids of contiguous E2 sequence, at least about 100 amino acids, at least about 200 amino acids, up to substantially all of the E2 protein.
  • a modified HCV E2 polypeptide is provided.
  • a polynucleotide encoding such a modified HCV E2 polypeptide is provided.
  • the polypeptide and/or the nucleic acid can be used in the formulation of a vaccine, e.g. a virus-like particle, a recombinant protein vaccine which can be formulated with an adjuvant, a vector vaccine, and the like.
  • a vaccine formulation comprising a polypeptide or a polynucleotide of the invention is provided.
  • an HCV provides for a partial or complete deletion of the HVR1 region.
  • the deletion may comprise some or all of amino acids 384-410 (for convenience, the numbering is shown in Figure 17).
  • the HCV E2“backbone” comprises E 2 sequence, amino acid residues 408-746, herein referenced as ⁇ HVR 408.
  • the HCV E2 backbone sequence comprises amino acid residues 411-746, herein referenced as ⁇ HVR1 411 .
  • the HVR1 sequence is not deleted, and the backbone comprises amino acid residues 384-746, herein referenced as sE2.
  • any of these backbone sequences may further comprise a deletion of the C-terminal region, amino acid residues 662-746.
  • a backbone HCV E2 polypeptide as defined above comprises one or both of the following modifications: (a) amino acid substitution N415P; (b) amino acid substitution N417P; (c) amino acid substitution A439P; (d) amino acid substitution H445P.
  • the amino acid substitution is one or both of N415P and H445P.
  • a backbone HCV E2 polypeptide as defined above, which is optionally combined with one or more amino acid substitutions that affect conformation described above, is modified to provide for insertion of a glycan.
  • Highly immunodominant epitopes that are associated with viral escape or non-neutralizing antibodies can be masked so as to focus an immune response to epitopes that are less immunodominant, but which are essential for virus entry and therefore are less likely to be altered in virus escape mutation and selection.
  • amino acid substitutions are made to generate a motif for N-glycosylation.
  • Such modification may comprise replacing residue Y632 with asparagine, i.e.
  • Y632N and introduction of a serine or threonine at G634, i.e. G634S or G634T.
  • R630 can be replaced with asparagine
  • R630N and Y632 can be replaced with serine or threonine, i.e. Y632S or Y632T.
  • Polypeptides incorporating these changes are expressed in a cell that provides for correct N-glycosylation, including without limitation mammalian cells.
  • modifications comprising: deletion of HVR1 region, introduction of one or more proline residues to stabilize confirmation, and introduction of residues to generate a motif for glycosylation, are all introduced in the HCV E2 polypeptide to generate an HCV E2 antigen.
  • one or more modifications from each group are introduced.
  • one or more modifications from a single group, from two groups, or from 3 groups are introduced.
  • the modified E2 polypeptide may further comprise one or more of the following amino acid changes: N417Q, N423Q, N448Q and N532Q, as described in published US Patent application 2015/0086580, herein specifically incorporated by reference. These mutations are to remove specific glycans near regions that elicit broadly neutralizing antibodies.
  • FIG. 1 Panels A-C. Location of antigenic domains on the HCV E2 envelope glycoprotein.
  • the E2 structure (PDB code 4MWF) is shown as a molecular surface shaded according to antigenic domains A-E (A: 581-584, 627-633; B: 431-439, 529-535; C: 544-549; D: 420-428, 441-443; E: 408-423).
  • FIG. 1 Energetic footprint mapping of epitope domain E.
  • a panel of antibodies was used to map the energetic footprint on E2 epitope domain E via alanine scanning mutagenesis.
  • Alanine substitution mutants were constructed in plasmids carrying the 1a H77C E1E2 coding sequence (GenBank accession nos. AF009606) as previously described. All the mutations were confirmed by DNA sequence analysis (Sequetech, Mountain View, CA) for the desired mutation and for exclusion of unexpected residue changes in the full-length E1E2 encoding sequence.
  • E2 mutant proteins were expressed in 293T cells and cell lysates were analyzed by ELISA.
  • FIG. 3 Energetic footprint mapping of epitope domain D.
  • a panel of antibodies (HC84.20, HC84.24, HC84.26) was used to map the energetic footprint on E2 epitope domain D via alanine scanning mutagenesis.
  • Alanine substitution mutants were constructed in plasmids carrying the 1a H77C E1E2 coding sequence (GenBank accession nos. AF009606) as previously described. All the mutations were confirmed by DNA sequence analysis (Sequetech, Mountain View, CA) for the desired mutation and for exclusion of unexpected residue changes in the full-length E1E2 encoding sequence.
  • E2 mutant proteins were expressed in 293T cells and cell lysates were analyzed by ELISA.
  • FIG. 4 Distinct epitope structures of domain E (aa 412-423).
  • the linear epitope from epitope domain E adopts an extended conformation when engaged by the HC33.1 human mAb (A) [12], versus the ⁇ hairpin conformation observed when bound to other antibodies including HCV1 (B), which was obtained via immunization of humanized mice.
  • FIG. 5 Computational analysis of E2 domain E residues for proline backbone conformation.
  • the HC33.1-E2 412-423 structure (PDB code 4XVJ) was analyzed by a Ramachandran plot analysis tool, which maps residue ⁇ backbone angle conformations onto amino acid backbone probability densities determined from a large set of x-ray crystal structures. This was used to identify positions suitable for proline substitution. The point corresponding to the N415 residue is labeled and shown as a blue square.
  • FIG. Structural models of N415P and H445P E2 proline mutants.
  • the structures of the HC33.1-E2 412-423 complex (A) and HC84.26AM-E2 434-446 complex (C) were used to generate models of the proline mutant N415P (B) and H445P (D) using the modeling program Rosetta.
  • FIG. 7 Binding of E2, ⁇ HVR1, and ⁇ HVR1 408 to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre-coated wells, and followed by incubation with each human mAb Human mAbat 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y-axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • FIG. 8 Binding of E2 ⁇ HVR1, ⁇ HVR1 N417T, and ⁇ HVR1 N415P to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre- coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y- axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • FIG. 9 Binding of E2, E2 ⁇ HVR1, ⁇ HVR1 N417T, and ⁇ HVR1 N415P to the AP33 epitope domain E mAb. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre-coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y-axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • FIG. 10 Binding of E2, N448Q, A439P, A440C/W616C to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre-coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y-axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the Sf2 genotype 1b strain.
  • O.D. optical density
  • FIG. 11 Binding of E2 H445P, ⁇ HVR1 408 H445P, and ⁇ HVR1 411 H445P to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre- coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y- axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • FIG. 12 Energetic footprint mapping of epitope domain A.
  • a panel of antibodies (CBH-4D, CBH-4G, CBH-4B, CBH-20, CBH-21, and CBH-22) was used to map the energetic footprint on E2 epitope domain A via alanine scanning mutagenesis.
  • Alanine substitution mutants were constructed in plasmids carrying the 1a H77C E1E2 coding sequence (GenBank accession nos. AF009606) as previously described. All the mutations were confirmed by DNA sequence analysis (Sequetech, Mountain View, CA) for the desired mutation and for exclusion of unexpected residue changes in the full-length E1E2 encoding sequence.
  • E2 mutant proteins were expressed in 293T cells and cell lysates were analyzed by ELISA.
  • FIG. 13 Structural basis for glycan-mediated antibody binding disruption in epitope domain A.
  • the E2 protein is shown using surface representation, with the region corresponding to epitope domain A.
  • the structure of a non-neutralizing antibody that binds this region (2A12) is shown for reference.
  • FIG. 14 Binding of ⁇ HVR1 F627N/V629T and ⁇ HVR1 R630N/Y632T to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre- coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y- axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • FIG. 15 Binding of E2 K628N/R630S, ⁇ HVR1 408 K628N/R630S, and ⁇ HVR1 411 K628N/R630S to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre-coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y-axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • FIG. 16 Binding of E2 Y632N/G634S, ⁇ HVR1 408 Y632N/G634S, and ⁇ HVR1 411 Y632N/G634S to a panel of E2 mAbs. Assays were performed with 2 ⁇ g of purified E2/ml that was captured by GNA pre-coated wells, and followed by incubation with each human mAb at 1 and 5 ⁇ g/ml (x-axis). Specific binding was detected by alkaline phosphatase-conjugated goat anti-human IgG. The y-axis shows the mean optical density (O.D.) values for triplicate wells, the mean of two experiments ⁇ SD. All E2 constructs were produced starting from the H77c strain.
  • O.D. optical density
  • “Flaviviridae virus” or“flavivirus” is meant any virus from the Flaviviridae family, including those viruses that infect humans and non-human animals.
  • the polynucleotide and polypeptides sequences encoding these viruses are well known in the art, and may be found at NCBI’s GenBank database, e.g., as Genbank Accession nos.
  • the term“flavivirus” includes any member of the family Flaviviridae, including, but not limited to, Dengue virus, including Dengue virus 1, Dengue virus 2, Dengue virus 3, Dengue virus 4 (see, e.g., GenBank Accession Nos. M23027, M19197, A34774, and M14931); Yellow Fever Virus; West Nile Virus; Japanese Encephalitis Virus; St. Louis Encephalitis Virus; Bovine Viral Diarrhea Virus (BVDV); and Hepatitis C Virus (HCV); and any serotype, strain, genotype, subtype, quasispecies, or isolate of any of the foregoing.
  • Dengue virus including Dengue virus 1, Dengue virus 2, Dengue virus 3, Dengue virus 4 (see, e.g., GenBank Accession Nos. M23027, M19197, A34774, and M14931); Yellow Fever Virus; West Nile Virus; Japanese Encephalitis Virus; St. Louis Encephalitis Virus; Bovine
  • the HCV is any of a number of genotypes, subtypes, or quasispecies, including, e.g., genotype 1, including 1a and 1b, 2, 3, 4, 6, etc. and subtypes (e.g., 2a, 2b, 3a, 4a, 4c, etc.), and quasispecies.
  • hepatitis C virus “HCV,” “non- A non-B hepatitis,” or “NANBH” are used interchangeably herein, and include any “genotype” or “subgenotype” (also termed “subtype”) of the virion, or portion thereof (e.g., a portion of the E2 protein of genotype Ia of HCV), that is encoded by the RNA of hepatitis C virus or that occurs by natural allelic variation.
  • the HCV genome comprises a 5'-untranslated region that is followed by an open reading frame (ORF) that codes for about 3,010 amino acids.
  • ORF open reading frame
  • the amino acids are subdivided into ten proteins in the order from 5' to 3' as follows: C; El; E2; NSl; NS2; NS3; NS4 (a and b); and NS5 (a and b). These proteins are formed from the cleavage of the larger polyprotein by both host and viral proteases.
  • the C, El, and E2 proteins are structural and the NS1-NS5 proteins are nonstructural proteins.
  • the C region codes for the core nucleocapsid protein.
  • El and E2 are glycosylated envelope proteins that coat the virus.
  • NS2 may be a zinc metalloproteinase.
  • NS3 is a helicase.
  • NS4a functions as a serine protease cofactor involved in cleavage between NS4b and NS5a.
  • NS5a is a serine phosphoprotein whose function is unknown.
  • the NS5b region has both RNA-dependent RNA polymerase and terminal transferase activity.
  • HCV genotypes There are about six major distinct HCV genotypes (e.g., genotypes 1, 2, 3, 4, 5, and 6) that are categorized by variations in the core protein and over 80 subgenotypes which exhibit further variation within each genotype, some of which include: Ia; Ib; Ic; 2a; 2b; 2c; 3a; 3b; 4a; 4b; 4c; 4d; 4e; 5a; and 6a.
  • the HCV RNA is directly translated into a continuous 3011-residue polypeptide chain.
  • a detailed annotation of the polyprotein, with the boundaries of cleavage products, may be found at Genbank, accession no. P27958, or AF009606, herein specifically incorporated by reference.
  • the polyprotein is subsequently cleaved to yield envelope and core proteins, which assemble into new virus particles, and enzymes essential for viral replication. At least ten proteins are encoded by the HCV genome. Both host cell and virally encoded proteases are required for maturation of the HCV polyprotein.
  • NS2 nonstructural protein 2
  • the NS2-mediated autoproteolysis generates the NS3 N terminus, and releases a 1984-residue polyprotein that incorporates a bifunctional 631-residue enzyme (NS3) possessing both serine protease and RNA helicase activities.
  • NS3 protease is responsible for processing the remaining polyprotein.
  • the amino acid sequence of the E2 protein which comprises residues 384-746 of the polyprotein for genotype 1, isolate 1a H77C (Genbank) is provided as SEQ ID NO:1, and modifications to the sequence are made with reference to SEQ ID NO:1. It will be understood by one of skill in the art that corresponding modifications are readily made in other HCV genotypes, by modifying the residue that corresponds to the named position in SEQ ID NO:1, and that for continuity with the literature, SEQ ID NO:1 may be considered to start with amino acid residue 384. In other words, for convenience in comparison to the literature, the proteins of the invention are numbered with respect to the HCV polyprotein, i.e. the starting residue of E2 is 384 and the ending residue is 746. Those of skill in the art will readily determine that numbering of these changes will vary depending on the selection of start site, and can easily be mapped to the reference sequences.
  • the antigenic protein of the invention is optionally truncated at the C-terminus, such that the protein comprises residues 384 to 746 (or 661).
  • the antigenic protein of the invention is optionally truncated at the amino terminus, such that the protein comprises residues 408 to 746 (or 408 to 661); or residues 411 to 746 (or residues 411 to 661).
  • the terms “neutralizes HCV,” “inhibits HCV,” and “blocks HCV” are used interchangeably to refer to the ability of an antibody of the invention to prevent HCV from infecting a given cell.
  • the term "effective dose” or “effective dosage” is defined as an amount sufficient to achieve or at least partially achieve the desired effect.
  • therapeutically effective dose is defined as an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. Amounts effective for this use will depend upon the severity of the disorder being treated and the general state of the patient's own immune system.
  • Polypeptide and “protein” as used interchangeably herein, can encompass peptides and oligopeptides. Where “polypeptide” is recited herein to refer to an amino acid sequence of a naturally-occurring protein molecule, "polypeptide” and like terms are not necessarily limited to the amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule, but instead can encompass biologically active variants or fragments, including polypeptides having substantial sequence similarity or sequence identify relative to the amino acid sequences provided herein. In general, fragments or variants retain a biological activity of the parent polypeptide from which their sequence is derived.
  • polypeptide refers to an amino acid sequence of a recombinant or non-recombinant polypeptide having an amino acid sequence of i) a native polypeptide, ii) a biologically active fragment of an polypeptide, or iii) a biologically active variant of an polypeptide.
  • Polypeptides suitable for use can be obtained from any species, e.g. , mammalian or non-mammalian (e.g. , reptiles, amphibians, avian (e.g.
  • polypeptides comprising a sequence of a human polypeptide are of particular interest.
  • the term "derived from” indicates molecule that is obtained directly from the indicated source (e.g., when a protein directly purified from a cell, the protein is “derived from” the cell) or information is obtained from the source, e.g. nucleotide or amino acid sequence, from which the molecule can be synthesized from materials other than the source of information.
  • isolated indicates that the recited material (e.g, polypeptide, nucleic acid, etc.) is substantially separated from, or enriched relative to, other materials with which it occurs in nature (e.g., in a cell).
  • a material e.g., polypeptide, nucleic acid, etc.
  • a material that is isolated constitutes at least about 0.1 %, at least about 0.5%, at least about 1 % or at least about 5% by weight of the total material of the same type (e.g., total protein, total nucleic acid) in a given sample.
  • subject and patient are used interchangeably herein to mean a member or members of any mammalian or non-mammalian species that may have a need for the pharmaceutical methods, compositions and treatments described herein.
  • Subjects and patients thus include, without limitation, primate (including humans), canine, feline, ungulate (e.g., equine, bovine, swine (e.g., pig)), avian, and other subjects.
  • Humans and non-human animals having commercial importance e.g. , livestock and domesticated animals are of particular interest.
  • subject and patient refer to a subject or patient susceptible to infection by a Flaviviridae virus, particularly HCV.
  • mammalian means a member or members of any mammalian species, and includes, by way of example, canines; felines; equines; bovines; ovines; rodentia, etc. and primates, particularly humans.
  • Non-human animal models, particularly mammals, e.g. primate, murine, lagomorpha, etc. may be used for experimental investigations.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for the novel unit dosage forms depend on the particular compound employed and the effect to be achieved, and the pharmacodynamics associated with each compound in the host.
  • a "pharmaceutically acceptable excipient,” “pharmaceutically acceptable diluent,” “pharmaceutically acceptable carrier,” and “pharmaceutically acceptable adjuvant” means an excipient, diluent, carrier, and adjuvant that are useful in preparing a pharmaceutical composition that are generally safe, non-toxic and neither biologically nor otherwise undesirable, and include an excipient, diluent, carrier, and adjuvant that are acceptable for veterinary use as well as human pharmaceutical use.
  • “A pharmaceutically acceptable excipient, diluent, carrier and adjuvant” as used in the specification and claims includes both one and more than one such excipient, diluent, carrier, and adjuvant.
  • a "pharmaceutical composition” is meant to encompass a composition suitable for administration to a subject, such as a mammal, especially a human.
  • a “pharmaceutical composition” is sterile, and is usually free of contaminants that are capable of eliciting an undesirable response within the subject (e.g., the compound(s) in the pharmaceutical composition is pharmaceutical grade).
  • Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal and the like.
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity.
  • Antibodies (Abs) and “immunoglobulins” (Igs) are glycoproteins having the same structural characteristics. While antibodies exhibit binding specificity to a specific antigen, immunoglobulins include both antibodies and other antibody-like molecules which lack antigen specificity. Polypeptides of the latter kind are, for example, produced at low levels by the lymph system and at increased levels by myelomas.
  • epitopic determinants means any antigenic determinant on an antigen to which the paratope of an antibody binds.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • conjugate is defined as a heterogeneous molecule formed by the covalent attachment of one or more polypeptide fragment(s) to one or more polymer molecule(s), wherein the heterogeneous molecule is water soluble, i.e. soluble in physiological fluids such as blood, and wherein the heterogeneous molecule is free of any structured aggregate.
  • a conjugate of interest is PEG.
  • structured aggregate refers to (1) any aggregate of molecules in aqueous solution having a spheroid or spheroid shell structure, such that the heterogeneous molecule is not in a micelle or other emulsion structure, and is not anchored to a lipid bilayer, vesicle or liposome; and (2) any aggregate of molecules in solid or insolubilized form, such as a chromatography bead matrix, that does not release the heterogeneous molecule into solution upon contact with an aqueous phase.
  • conjugate encompasses the aforementioned heterogeneous molecule in a precipitate, sediment, bioerodible matrix or other solid capable of releasing the heterogeneous molecule into aqueous solution upon hydration of the solid.
  • mAb monoclonal antibody
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site. Each mAb is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they can be synthesized by cell culture, uncontaminated by other immunoglobulins.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made in an immortalized B cell or hybridoma thereof, may be made by recombinant DNA methods, including without limitation yeast display.
  • label when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the antibody.
  • the label may itself be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • solid phase is meant a non-aqueous matrix to which the antibody of the present invention can adhere.
  • solid phases encompassed herein include those formed partially or entirely of glass (e.g. controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol and silicones.
  • the solid phase can comprise the well of an assay plate; in others it is a purification column (e.g. an affinity chromatography column). This term also includes a discontinuous solid phase of discrete particles, such as those described in U.S. Pat. No. 4,275,149.
  • vaccine as used herein, is meant a composition; a formulation comprising a modified polypeptide of the invention; a virus or virus-like particle comprising a modified polypeptide of the invention complex; or a DNA encoding a modified polypeptide of the invention complex, which, when administered to a subject, induces cellular or humoral immune responses as described herein.
  • Some embodiments of the invention provide a method of stimulating an immune response in a mammal, which can be a human or a preclinical model for human disease, e.g. mouse, ape, monkey etc.
  • Stimulating an immune response includes, but is not limited to, inducing a therapeutic or prophylactic effect that is mediated by the immune system of the mammal. More specifically, stimulating an immune response in the context of the invention refers to eliciting cellular or humoral immune responses, thereby inducing downstream effects such as production of antibodies, antibody heavy chain class switching, maturation of APCs, and stimulation of cytolytic T cells, T helper cells and both T and B memory cells.
  • vaccine compositions are suitably formulated to be compatible with the intended route of administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as
  • the pH of the composition can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • Systemic administration of the composition is also suitably accomplished by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • Vaccine compositions may include an aqueous medium, pharmaceutically acceptable inert excipient such as lactose, starch, calcium carbonate, and sodium citrate.
  • Vaccine compositions may also include an adjuvant, for example Freud's adjuvant.
  • Vaccines may be administered alone or in combination with a physiologically acceptable vehicle that is suitable for administration to humans.
  • Vaccines may be delivered orally, parenterally, intramuscularly, intranasally or intravenously. Oral delivery may encompass, for example, adding the compositions to the feed or drink of the mammals.
  • Factors bearing on the vaccine dosage include, for example, the weight and age of the mammal.
  • compositions for parenteral or intravenous delivery may also include emulsifying or suspending agents or diluents to control the delivery and dose amount of the vaccine.
  • modified polypeptides of the invention and polynucleotides that encode such modified polypeptides can be used in various HCV vaccine formulations known in the art, as a substitution for the wild-type HCV E2 sequence.
  • HCV vaccines include, without limitation, formulation of isolate polypeptides, e.g. E2 alone or in combination with E1 as separate molecules or as heterodimeric E1E2, and an adjuvant.
  • a protein complex of HCV proteins, including E2 of the present invention can be formulated with T-cell adjuvant immunostimulating complex matrix (IMX).
  • IMX T-cell adjuvant immunostimulating complex matrix
  • the polypeptides of the invention can be fragmented to generate a peptide vaccine, e.g. administered with poly-L- arginine, can be formulated as a vaccine.
  • Polynucleotides encoding the modified polypeptides of the invention can be administered in plasmid form, in a virus genome, including adenovirus, alphaviruses, canary pox, ovine atadenovirus and semliki-like viral particles.
  • Advances in molecular virology have enabled the manipulation of viruses for delivery of foreign genetic material to mammalian cells.
  • Their highly evolved mechanisms for cell entry and gene expression within the host cell remain intact and viral vectors can be rendered non-pathogenic and non-replicative by deletions at specific locus.
  • the polypeptides of the invention are formulated for vaccine delivery as virus-like particles (VLPs).
  • VLPs virus-like particles
  • Ad vectors are the best characterized viral vectors and have emerged as the most potent at T-cell priming in non-human primates (NHPs) and humans.
  • Ad-based vaccines are particularly attractive gene vehicles as they can stably express large foreign inserts ( ⁇ 10 kbp), they remain epichromosomal and can be easily rendered replication defective by deletion of the E1 locus.
  • the application discloses herein a modified HCV E2 polypeptide, which is altered from the wild-type in various ways to increase desired immune responses, which generate neutralizing antibodies across multiple HCV genotypes; and the reduce undesirable immune responses that are readily avoided by escape mechanisms.
  • modified polypeptides find use in screening assays, generation of monoclonal antibodies, and in vaccines.
  • HCV E2 protein is provided, e.g. as the full-length E2 protein, or a modified peptide derived therefrom, including peptides derived from residues 408-746, residues 411-746, residues 384-746 of HCV E2 protein, where the epitope is of sufficient length to provide for binding specificity substantially similar to the specificity of binding to the native protein, e.g.
  • Peptides can be produced using techniques well known in the art. Such techniques include chemical and biochemical synthesis. Examples of techniques for chemical synthesis of peptides are provided in Vincent, in Peptide and Protein Drug Delivery, New York, N.Y., Dekker, 1990. Examples of techniques for biochemical synthesis involving the introduction of a nucleic acid into a cell and expression of nucleic acids are provided in Ausubel, Current Protocols in Molecular Biology, John Wiley, and Sambrook, et al in Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, 1989.
  • HCV E2 polypeptides of the invention comprise one or more of the following modifications: deletion of all or part of the HVR1 region; one or more of (a) amino acid substitution N415P; (b) amino acid substitution N417P; (c) amino acid substitution A439P; (d) amino acid substitution H445P; and amino acid substitutions are made to generate a motif for N-glycosylation.
  • modification may comprise replacing residue Y632 with asparagine, i.e. Y632N and introduction of a serine or threonine at G634, i.e. G634S or G634T.
  • R630 can be replaced with asparagine
  • R630N and Y632 can be replaced with serine or threonine, i.e. Y632S or Y632T.
  • Polypeptides incorporating these changes are expressed in a cell that provides for correct N-glycosylation, including without limitation mammalian cells.
  • the polypeptide may optionally comprise one or more of the following amino acid substitutions: N417Q, N423Q, N448Q and N532Q. These mutations are to remove specific glycans near regions that elicit broadly neutralizing antibodies.
  • the invention also provides isolated nucleic acids encoding the modified HCV E2 polypeptide, vectors and host cells comprising the nucleic acid, and recombinant techniques for the production of the modified polypeptide. As is known in the art, various polynucleotides can be devised with respect to codon usage to produce a desired polypeptide, and one of skill in the art can readily generate a polynucleotide sequence that encodes a modified E2 protein.
  • sequence of isolates 1a H77C (Genbank AF009606) and 1bSF (Genbank JN118490) can be used, without limitation.
  • a contiguous nucleotide sequence is at least about 20 nt., at least about 25 nt, at least about 50 nt., at least about 75 nt, at least about 100 nt, and up to the complete coding sequence may be used.
  • the nucleic acid encoding it is inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
  • DNA encoding the modified polypeptide is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Many vectors are available.
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • the modified polypeptides of this invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous or homologous polypeptide, which include a signal sequence or other polypeptide having a specific cleavage site at the N- terminus of the mature protein or polypeptide, and the like.
  • a heterologous signal sequence selected preferably may be one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.
  • the signal sequence is substituted by a prokaryotic signal sequence selected.
  • An "isolated" nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the antibody nucleic acid.
  • An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the nucleic acid molecule as it exists in natural cells.
  • an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the antibody where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • control sequences refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase.
  • enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • the expressions "cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • Suitable host cells for cloning or expressing the DNA are the prokaryote, yeast, or higher eukaryote cells.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR(CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod.
  • monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TR1 cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1.982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
  • Host cells are transformed with the above-described expression or cloning vectors for modified polypeptide production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the modified polypeptide composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, and the like as known in the art.
  • antibodies against E2 protein can be used as affinity reagents for purification.
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the modified polypeptide(s) are used in a screening method to select for antibodies optimized for affinity, specificity, and the like.
  • variable regions will initially comprise one or more of the provided CDR sequences, e.g. a framework variable region comprising CDR1, CDR2, CDR3 from the heavy and light chain sequences.
  • CDR sequences e.g. a framework variable region comprising CDR1, CDR2, CDR3 from the heavy and light chain sequences.
  • the modified polypeptide of the invention is used as an immunogen, including without limitation vaccine preparation. Methods of Use
  • an immunologically effective amount of one or more modified polypeptides of the invention which may be conjugated to a suitable carrier molecule, polynucleotides encoding such modified polypeptides, including viral vectors, are administered to a patient by administrations of a vaccine, in a manner effective to result in an improvement in the patient's condition. successive, spaced
  • VLPs virus-like particles
  • examples of VLPs used as peptide carriers are hepatitis B virus surface antigen and core antigen, hepatitis E virus particles, polyoma virus, bovine papilloma virus, and the like.
  • modified polypeptides of the invention are coupled to one of a number of carrier molecules, known to those of skill in the art.
  • a carrier protein must be of sufficient size for the immune system of the subject to which it is administered to recognize its foreign nature and develop antibodies to it.
  • the carrier molecule is directly coupled to the immunogenic peptide.
  • the coupling reaction may require a free sulfhydryl group on the peptide.
  • an N-terminal cysteine residue is added to the peptide when the peptide is synthesized.
  • traditional succinimide chemistry is used to link the peptide to a carrier protein. Methods for preparing such peptidexarrier protein conjugates are generally known to those of skill in the art and reagents for such methods are commercially available (e.g., from Sigma Chemical Co.). Generally about 5-30 peptide molecules are conjugated per molecule of carrier protein.
  • Exemplary carrier molecules include proteins such as keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA), flagellin, influenza subunit proteins, tetanus toxoid (TT), diphtheria toxoid (DT), cholera toxoid (CT), a variety of bacterial heat shock proteins, glutathione reductase (GST), or natural proteins such as thyroglobulin, and the like.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • flagellin influenza subunit proteins
  • influenza subunit proteins tetanus toxoid
  • TT tetanus toxoid
  • DT diphtheria toxoid
  • CT cholera toxoid
  • GST glutathione reductase
  • natural proteins such as thyroglobulin, and the like.
  • the carrier molecule is a non-protein, such as Ficoll 70 or
  • a peptide vaccine composition may comprise single or multiple copies of the same or different modified polypeptide of the invention.
  • the peptide vaccine composition may contain different immunogenic peptides with or without flanking sequences, combined sequentially into a polypeptide and coupled to the same carrier.
  • immunogenic peptides may be coupled individually as peptides to the same or a different carrier, and the resulting immunogenic peptide-carrier conjugates blended together to form a single composition, or administered individually at the same or different times.
  • peptide vaccine compositions are administered with a vehicle.
  • vehicle The purpose of the vehicle is to emulsify the vaccine preparation.
  • Numerous vehicles are known to those of skill in the art, and any vehicle which functions as an effective emulsifying agent finds utility in the present invention.
  • an immunological adjuvant may be included in the vaccine formulation.
  • Exemplary adjuvants known to those of skill in the art include water/oil emulsions, non-ionic copolymer adjuvants, e.g., CRL 1005 (Optivax; Vaxcel Inc., Norcross, Ga.), aluminum phosphate, aluminum hydroxide, aqueous suspensions of aluminum and magnesium hydroxides, bacterial endotoxins, polynucleotides, polyelectrolytes, lipophilic adjuvants and synthetic muramyl dipeptide (norMDP) analogs.
  • CRL 1005 Optivax; Vaxcel Inc., Norcross, Ga.
  • aluminum phosphate aluminum hydroxide
  • aqueous suspensions of aluminum and magnesium hydroxides aqueous suspensions of aluminum and magnesium hydroxides
  • bacterial endotoxins polynucleotides
  • polyelectrolytes polyelectrolytes
  • lipophilic adjuvants and synthetic muramyl dipeptide (norMDP) analogs.
  • Suitable pharmaceutically acceptable carriers for use in an immunogenic proteinaceous composition of the invention are well known to those of skill in the art.
  • Such carriers include, for example, phosphate buffered saline, or any physiologically compatible medium, suitable for introducing the vaccine into a subject.
  • Controlled release preparations may be achieved by the use of polymers to complex or absorb the peptides or antibodies. Controlled delivery may accomplished using macromolecules such as, polyesters, polyamino acids, polyvinyl pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate, the concentration of which can alter the rate of release of the peptide vaccine.
  • the peptides may be incorporated into polymeric particles composed of e.g., polyesters, polyamino acids, hydrogels, polylactic acid, or ethylene vinylacetate copolymers.
  • the peptide vaccine is entrapped in microcapsules, liposomes, albumin microspheres, microemulsions, nanoparticles, nanocapsules, or macroemulsions, using methods generally known to those of skill in the art.
  • the vaccine is a vector.
  • a“vector” is a tool that allows or facilitates the transfer of an entity from one environment to another.
  • WO 99/60164 and WO98/00166 van Ginkel et al.,“Adenoviral gene delivery elicits distinct pulmonary-associated T helper cell responses to the vector and to its transgene,” J Immunol 159(2):685-93, 1997; and Osterhaus et al,“Vaccination against acute respiratory virus infections and measles in man,” Immunobiology 184(2-3):180-92, 1992, which contain information concerning expressed gene products, antibodies and uses thereof, vectors for in vivo and in vitro expression of exogenous nucleic acid molecules, promoters for driving expression or for operatively linking to nucleic acid molecules to be expressed, method and documents for producing such vectors, compositions comprising such vectors or nucleic acid molecules or antibodies, dosages, and modes and/or routes of administration (including compositions for nasal administration), inter alia, can be employed in the practice of this invention and are incorporated by herein reference in their entireties.
  • the vector can be a viral vector, a bacterial vector, a protozoan vector, a retrotransposon, a transposon, a virus shell, or a DNA vector.
  • the viral vector, the bacterial vector, the protozoan vector and the DNA vector can be recombinant vectors.
  • the vector can be an adenovirus.
  • the adenovirus recombinant can include E1-defective, E3-defective, and/or E4-defective adenovirus vectors, or the“gutless” adenovirus vector, where all viral genes are deleted.
  • adenovirus recombinant is constructed by cloning specific transgenes or fragments of transgenes into any of the adenovirus vectors such as those described above.
  • the adenovirus recombinant can be used to transduce epidermal or mucosal cells of a subject in a noninvasive mode for use as an immunizing agent.
  • the adenovirus vector can be defective in its E1 region.
  • the adenovirus vector can be defective in its E3 region.
  • the adenovirus vector can be defective in its E1 and E3 regions.
  • the DNA is in plasmid form.
  • a vaccine of the present invention can be administered to patient by different routes such as intravenous, intraperitoneal, subcutaneous, intramuscular, or orally.
  • a preferred route is intramuscular or oral.
  • Suitable dosing regimens are preferably determined taking into account factors well known in the art including age, weight, sex and medical condition of the subject; the route of administration; the desired effect; and the particular conjugate employed (e.g., the peptide, the peptide loading on the carrier, etc.).
  • the vaccine can be used in multi-dose vaccination formats.
  • the timing of doses depends upon factors well known in the art. After the initial administration one or more booster doses may subsequently be administered to maintain antibody titers.
  • An example of a dosing regimen would be a dose on day 1, a second dose at or 2 months, a third dose at either 4, 6 or 12 months, and additional booster doses at distant times as needed.
  • the invention provides a means for classifying the immune response to peptide vaccine, e.g., 9 to 15 weeks after administration of the vaccine; by measuring the level of antibodies against the immunogenic peptide of the vaccine.
  • the vaccine formulations of the present invention may be used in immunization for the various HCV associated diseases.
  • the recipient is at a high risk of infection.
  • the vaccine formulation is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the vaccine formulation is suitably administered by pulse infusion, particularly with declining doses of the vaccine.
  • the appropriate dosage of vaccine will depend on the type of disease to be treated, the severity and course of the disease, whether the vaccine is administered for preventive purposes, previous therapy, the patient's clinical history and response to the vaccine, and the discretion of the attending physician.
  • the vaccine is suitably administered to the patient at one time or over a series of treatments.
  • an article of manufacture containing materials useful for the vaccination described above comprises a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agent in the composition is one or more antibodies in a formulation of the invention as described above.
  • the label on, or associated with, the container indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a pharmaceutically-acceptable buffer such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • Therapeutic formulations are prepared for storage by mixing the vaccine having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • the vaccine composition will be formulated, dosed, and administered in a fashion consistent with good medical practice.
  • the "therapeutically effective amount" of the vaccine to be administered will be governed by clinical considerations, and is the minimum amount necessary to reduce virus titer in an infected individual.
  • An immunologically effective dose is one that stimulates the immune system of the patient to establish a level immunological memory sufficient to provide long term protection against disease caused by infection with HCV. More precise dosages should be determined by assessing the immunogenicity of the vaccine produced so that an immunologically effective dose is delivered.
  • the therapeutic dose may be at least about 0.01 ⁇ g/kg body weight, at least about 0.05 ⁇ g/kg body weight; at least about 0.1 ⁇ g/kg body weight, at least about 0.5 ⁇ g/kg body weight, at least about 1 ⁇ g/kg body weight, at least about 2.5 ⁇ g/kg body weight, at least about 5 ⁇ g/kg body weight, and not more than about 100 ⁇ g/kg body weight. It will be understood by one of skill in the art that such guidelines will be adjusted for the molecular weight of the active agent, e.g. in the use of vaccine fragments, or in the use of vaccine conjugates.
  • the dosage may also be varied for localized administration, or for systemic administration, e.g. i.m., i.p., i.v., and the like.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, his
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • the formulation is an aqueous suspension
  • such can contain the active agent in a mixture with a suitable excipient(s).
  • excipients can be, as appropriate, suspending agents (e.g., sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia); dispersing or wetting agents; preservatives; coloring agents; and/or flavoring agents.
  • Suppositories e.g., for rectal administration of agents, can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter and polyethylene glycols.
  • HC33.1 which was isolated from an HCV- infected blood donor, can neutralize virus bearing the E2 Asn417Ser and Asn417Thr adaptive mutations with the glycosylation shift to Asn415. Moreover, infectious HCV virions containing the Asn417Ser mutation displayed increased sensitivity to neutralization by HC33.1 and related antibodies, suggesting that the glycosylation shift actually enhanced binding. To understand how HC33.1 is able to penetrate the glycan shield of HCV created by this shift, we determined the crystal structure of this broadly neutralizing human mAb in complex with its E2412–423 epitope.
  • HVR1 hypervariable region 1
  • A–E hypervariable region 1
  • Antigenic domain A is associated with non-neutralizing antibodies and constitutes another major decoy.
  • some antigenic domain B epitopes, and most D and E epitopes elicit antibodies that are broadly neutralizing among the major HCV genotypes and subtypes.
  • Antibodies to domain C epitopes neutralize HCV with more restricted genotype and subtype profiles.
  • Antigenic domain E is composed of overlapping linear epitopes located within amino acids 412–423 (QLINTNGSWHIN) of E2 (E2412– 423). This region is involved in HCV binding to the CD81 entry receptor, which likely explains why it is so highly conserved among >5,500 E2 sequences in the GenBank database. Accordingly, antibodies to E2412–423 hold considerable promise for vaccine development. Broadly neutralizing mAbs targeting this region have been isolated from immunized mice and HCV-infected individuals. These antibodies, which block the interaction of E2 with CD81, include the rodent mAbs AP33 and 3/11, and human mAbs HCV1 and HC33.1.
  • the HC33.1 antibody was expressed as a single-chain Fv fragment (scFv) by in vitro folding from inclusion bodies produced in Escherichia coli.
  • the scFv construct consisted of the heavy chain variable (VH) region (residues Glu1–Ser127) connected to the light chain variable (VL) region (residues Gln1–Leu110) by an 18-residue linker (GSTGGGGSGGGGSGGGGS).
  • VH heavy chain variable
  • VL light chain variable
  • the HC33.1 scFv was cloned into the expression vector pET- 26b (Novagen) and expressed as inclusion bodies in BL21(DE3) E.
  • Bacteria were grown at 37oC in LB medium to an absorbance of 0.6–0.8 at 600 nm, and induced with 1 mM isopropyl- ⁇ -D-thiogalactoside. After incubation for 3 h, the bacteria were harvested by centrifugation and resuspended in 50 mM Tris- HCl (pH 8.0) containing 0.1 M NaCl and 2 mM EDTA; cells were disrupted by sonication.
  • Tris- HCl pH 8.0
  • Inclusion bodies were washed extensively with 50 mM Tris-HCl (pH 8.0) and 2% (v/v) Triton X- 100, then dissolved in 8 M urea, 50 mM Tris-HCl (pH 8.0), and 10 mM DTT.
  • inclusion bodies were diluted into ice-cold folding buffer containing 1 M L-arginine-HCl, 50 mM Tris-HCl (pH 8.0), 1 mM EDTA, 3 mM reduced glutathione, and 0.9 mM oxidized glutathione, to a final protein concentration of 60 mg/l. After 72 h at 4°C, the folding mixture was concentrated 50-fold, dialyzed against 50 mM MES (pH 6.0), and centrifuged to remove aggregates. Correctly folded HC33.1 scFv was then purified using sequential Superdex 75 HR and MonoQ columns (GE Healthcare).
  • X-ray diffraction data were collected in- house at 100 K using a Rigaku R-AXIS IV++ area detector.
  • the HC33.1–E2412–423 crystal belongs to space group P212121 with one complex molecule per asymmetric unit.
  • Diffraction data were indexed, integrated, and scaled with the program CrystalClear (Rigaku).
  • Data collection statistics are shown in Table 1.
  • Modeling of Epitope Mutants and Glycans–Rosetta version 2.3 was used to perform a computational alanine scan and escape mutant simulations of the E2412–423 epitope using the interface mutagenesis module, removing the N-terminal arginine residue from the HC33.1– E2412–423 complex structure prior to modeling to avoid any influence of non-epitope residues on results.
  • Extra side chain rotamers were included to ensure sufficient sampling (“-ex1–ex2– ex3”), and a separate simulation for non-alanine mutants was performed incorporating backbone and off- rotamer side chain minimization of the complex before and after mutation (“- min_interface–int_bb–int_chi”).
  • Glycans were modeled using the GlyProt web server, using oligomannose glycans as high mannose glycans were found to be predominant in native E2. As with the Rosetta simulations, the engineered N terminal arginine residue was removed from the peptide in the complex structure prior to input into the server.
  • residues 414–415 of the peptide form an anti-parallel ⁇ -sheet with ⁇ -strand F of the VH domain, while the rest of the peptide adopts a coil conformation.
  • these residues form part of the first strand of the ⁇ -hairpin.
  • the N- terminal coil preceding the ⁇ -strand of the bound peptide extends behind the FG ⁇ -sheet of the HC33.1 VH domain.
  • the peptide makes a turn at residues 416–419, surrounding the F strand of VH, while its C-terminus sits loosely in front of the FG ⁇ -sheet.
  • the peptide interacts predominantly with the HC33.1 H chain, which mediates 90% of the 186 total contacts. Interactions with the L chain are restricted to the side chains of Leu413, Trp420 and His421, at the N- and C-termini of E2412–423.
  • HC33.1 has an unusually long VH complementarity- determining region 3 (CDR3) (22 residues compared to 14 and 18 residues for AP33 and HCV1, respectively).
  • the C-terminus of strand F and N terminus of strand G which constitute part of this VHCDR3, are each 3–4 residues longer in HC33.1 than in HCV1 or AP33.
  • the extended F strand permits E2412–423 to“clip” onto the antibody through formation of the anti- parallel ⁇ -sheet described above.
  • VHCDR1 and VHCDR2 mainly contact the ⁇ -turn of the bound peptide.
  • E2412–423 in addition to the existence of the ⁇ -hairpin in the majority of mAb-bound structures, E2412–423 likely adopts a ⁇ - hairpin conformation in the E2 protein, and maintains a similar structure after binding to AP33 or HCV1. However, this ⁇ -hairpin structure can be disrupted through interactions with certain antibodies, such as 3/11 or HC33.1.
  • the HC33.1–E2412–423 Interface The HC33.1–E2412–423 complex buries a total solvent accessible surface area of 1744 ⁇ 2, with 11 peptide residues contacting 23 antibody residues.
  • Sc shape correlation statistic
  • the HC33.1–E2412–423 interface may be subdivided into three regions, according to the distribution of contacting residues on the peptide.
  • N-terminal residues 412–416 interact exclusively with VHCDR3, except for Leu413, which also contacts VLCDRI Tyr33 and VLCDR2 Asp51 .
  • Region 2 comprising residues 417-419 (NGS) at the peptide ⁇ - turn, interacts with VHCDRI and VHCDR2.
  • C-terminal residues 420-423 contact HC33.1 over a broader area that includes 1 1 residues from VLCDR3, VH framework region 2 (VHFR2), VHCDR2, VHFR3, and VHCDR3.
  • Trp420 makes two main-chain–sidechain hydrogen bonds with HC33.1: Trp420 N–O ⁇ VHCDR2 Ser52 and Trp420 O–N ⁇ VHCDR3 Lys112.
  • Trp420 Gly418 is completely buried in the HC33.1–E2412–423 interface, where it makes 21 close contacts with the backbones of VHCDR1 Asn31 and Phe32, and of VHCDR2 Ser52, Ser53 and Ser54.
  • HC33.1 ability of HC33.1 to accommodate the glycosylation shift to Asn415 is explained by the unique conformation of E2412–423 induced (or captured) by this mAb, rather than by HC33.1 binding in a different way to the ⁇ -hairpin or extended conformation of E2412–423 recognized by HCV1, AP33 or 3/11.
  • the buried Asn415 residue is part of the first strand of the E2412– 423 ⁇ - hairpin, whereas in the HC33.1–E2412–423 complex, Asn415 is part of a strand that forms an anti-parallel ⁇ -sheet with strand F of VH, such that the Asn415 side chain flanks the antibody and is 58% solvent-accessible.
  • a glycan chain can be attached to the side-chain amide of Asn415, based on modeling using the GlyProt server, indicating that glycosylation of E2 at Asn415 would not prevent binding of HC33.1.
  • HC33.1 binding to the flexible E2412–423 epitope induces a conformation that allows accommodation of multiple glycans in the interface, whether at Asn417 and Asn423 in wild-type E2, or at Asn415 and Asn423 in glycosylation-shifted E2 mutants.
  • HCV may employ structural flexibility as an immune evasion strategy.
  • the crystal structure of the HCV E2 core revealed that ⁇ 60% of all residues are either disordered or in loops, implying considerable overall flexibility.
  • Neutralizing and non-neutralizing mouse mAbs specific for an E2 epitope comprising residues 427–446 were found to bind distinct conformations of this epitope that determined recognition specificity.
  • the E2412–423 epitope can adopt at least three different conformations, which may contribute to reducing its immunogenicity in HCV-infected individuals.
  • the HC33.1–E2412–423 structure explains the ability of HC33.1 to accommodate glycans at Asn417 and Asn423 in wild-type E2, or at Asn415 and Asn423 in glycosylation-shifted E2 mutants.
  • a possible additional glycan has been proposed for the mutation Ser419Asn, which was observed in the context of other mutations, notably Asn417Thr, in a cell-based study of HC33.1 escape mutants.
  • the Ser419Asn and Asn417Thr mutations may generate a new glycan at position 419 due to creation of a Thr-X-Asn glycosylation sequon.
  • Thr-X-Asn sequon is one of several reported alternative motifs for N-glycosylation.
  • Ser419Asn and Asn417Thr permitted modeling of a glycan at Asn419.
  • this glycan if present, is not predicted to abolish antibody binding completely.
  • Asn417Ser/Ser419Asn escape variants could still be neutralized by HC33.1 , albeit at higher antibody concentrations, and HC33.1 retained significant binding to recombinant E1 E2 bearing these mutations.
  • a glycan at Asn419 may influence the structure or conformational dynamics of the E2412-423 epitope, as noted in studies of peptide dynamics, or interact with other regions of E2.
  • HCV is an RNA virus in the flavivirus family, with a 9.5 kilobase genome, six distinct genotypes and several subtypes. Even within subtypes, there is remarkable sequence variability, particularly within envelope proteins E1 and E2, which is thought to result from mutation of surface residues to evade immune response in the host. Given its high sequence variability and resultant lack of amenability to targeted approaches, HCV is often compared with HIV, which historically has garnered greater attention from the research community. A recent phase 2 clinical trial of a monoclonal antibody targeting an epitope of HCV E2 demonstrated that HCV actively escapes the immune response via mutation when under immune pressure (Babcock et al. PLoS One 2014, 9(6):e100325).
  • a vaccine for HCV must overcome the high sequence variability of this virus, and its capacity to escape, by inducing protective antibodies that target conserved epitopes.
  • the recently described structures of HCV E2 (the primary target of the human antibody response to HCV), and antibodies bound to several E2 epitopes (see, for example, Krey et al. PLoS Pathog 2013, 9(5):e1003364; Meola et al. J Virol 2015, 89(4):2170-2181; Li et al. J Biol Chem 2015, 290(16):10117-10125; Pantua et al. J Mol Biol 2013, 425(11):1899-1914), in conjunction with advanced bioinformatics and structural modeling algorithms, permit the rational structure-based design of optimized variants of the E2 protein for use in an effective vaccine.
  • the present invention describes these designs.
  • H77c E2 (GenBank accession no. AF009606), amino acid (aa) 384 to 661, 411-661 or 408-661 was cloned into the expression vector pSec in-frame with the Ig ⁇ signal peptide sequence and fused with a myc and six-histidine tag at the carboxyl terminus. Amino acids substitution and glycan insertion at specific designed residues were created by using a QuikChange II site-directed mutagenesis kit (Agilent, La Jolla, CA) in accordance with the manufacturer's instructions. Mutations were confirmed by DNA sequence analysis (ElimBiopharm, Hayward, CA).
  • ELISA Immunoassays As described previously microtiter plates were prepared by coating each well with 500 ng of GNA and blocking with 2.5% non-fat dry milk and 2.5% normal goat serum. Purified wt HCV H77c E2 or mutant E2, were captured by GNA onto the plate and later bound by 1 and 5 ⁇ g/ml of human mAb. The bound human mAb was detected by incubation with alkaline phosphatase-conjugated goat anti-human IgG followed by incubation with p-nitrophenyl phosphate for color development. Absorbance was measured at 405 nm and 570 nm.
  • thermodynamic parameters for the binding of E2 domain E peptide (aa 412-423) and proline mutants to the HC33.1 antibody were carried out using a MicroCal ITC200 titration microcalorimeter.
  • Purified HC33.1 antibody was exhaustively dialyzed against PBS buffer. In a typical experiment, 2 ⁇ l aliquots of 100–500 ⁇ M peptide solution were injected from a 40 ⁇ l rotating syringe into the sample cell containing 200 ⁇ l of 10–30 ⁇ M HC33.1 antibody solution at 25 °C. For each titration experiment, an identical buffer dilution correction was conducted; these heats of dilution were subtracted from the corresponding binding experiment.
  • HC84.26AM-E2 434-446 Structure Determination - Protein Expression and Purification
  • the HC84.26AM antibody was expressed as a single-chain Fv fragment (scFv) by in vitro folding from inclusion bodies produced in Escherichia coli.
  • the scFv construct consisted of the heavy chain variable (V H ) region (residues Glu1–Ser127) connected to the light chain variable (V L ) region (residues Gln1–Leu110) by an 18-residue linker (GSTGGGGSGGGGSGGGGS).
  • the HC84.26AM scFv was cloned into the expression vector pET-26b (Novagen) and expressed as inclusion bodies in BL21(DE3) E. coli cells (Novagen). Bacteria were grown at 37 o C in LB medium to an absorbance of 0.6–0.8 at 600 nm, and induced with 1 mM isopropyl- ⁇ -D- thiogalactoside. After incubation for 3 h, the bacteria were harvested by centrifugation and resuspended in 50 mM Tris-HCl (pH 8.0) containing 0.1 M NaCl and 2 mM EDTA; cells were disrupted by sonication.
  • Tris-HCl pH 8.0
  • Inclusion bodies were washed extensively with 50 mM Tris-HCl (pH 8.0) and 2% (v/v) Triton X-100, then dissolved in 8 M urea, 50 mM Tris-HCl (pH 8.0), and 10 mM DTT.
  • inclusion bodies were diluted into ice-cold folding buffer containing 1 M L-arginine-HCl, 50 mM Tris-HCl (pH 8.0), 1 mM EDTA, 3 mM reduced glutathione, and 0.9 mM oxidized glutathione, to a final protein concentration of 60 mg/l.
  • the folding mixture was concentrated 50-fold, dialyzed against 50 mM MES (pH 6.0), and centrifuged to remove aggregates. Correctly folded HC84.26AM scFv was then purified using sequential Superdex 75 HR and MonoQ columns (GE Healthcare).
  • HC84.26AM (10 mg/ml) was mixed with E2 434–446 peptide (NTGWLAGLFYQHK) (GenScript) in a 1:5 molar ratio. Crystals were grown at room temperature in sitting drops containing 1 ⁇ l of the complex solution mixed with 1 ⁇ l of reservoir solution consisting of 1.6 M sodium phosphate monobasic, 0.4 M potassium phosphate dibasic, and 0.1 M sodium phosphate dibasic/citric acid (pH 4.2).
  • a panel of human monoclonal antibodies (mAbs) targeting the HCV E2 protein has been isolated and characterized Keck et al. J Virol 2013, 87(1):37-51.) Based on cross-competition studies, epitope mapping, as well as structural studies (Krey et al. PLoS Pathog 2013, 9(5):e1003364; Meola et al. J Virol 2015, 89(4):2170-2181; Li et al. J Biol Chem 2015, 290(16):10117-10125), these antibodies have been found to target several regions of the E2 protein surface, referred to as antigenic domains A-E (Table 1, Figure 1). While antigenic domains B, D, and E are associated with neutralizing antibodies, due at least in part due to overlap with the CD81 binding site critical for human cell entry, antigenic domain A is not associated with neutralizing antibodies.
  • the conformation strategy of antigen design entails producing selected mutants near an epitope site, improving its chance of antibody recognition and thus antibody induction by locking the structure into an antibody-bound conformation. Selected mutants will not include key binding (“hotspot”) residues as such mutants would likely reduce antibody binding and/or alter the antigenic specificity of antibodies induced to the site. Hotspot residues were identified via alanine scanning (reducing mAb binding to ⁇ 20% compared to wild-type affinity), as shown for antigenic domains E and D ( Figures 2, 3). Such designs utilize the residue proline to lock the backbone conformation of residues proximal to the epitope.
  • antigenic domain E (aa 412-423), which based on x-ray structural characterization adopts several widely varying conformations depending on the mAb to which it is bound ( Figure 4).
  • mAb HC33.1 recognizes the antigen domain E epitope in a specific extended conformation ( Figure 4A), and is not susceptible to the glycan shift to residue N415 which allows viral escape from antibodies that target the ⁇ -hairpin conformation of this epitope ( Figure 4B). This is evident from the alanine scanning data which indicate that N415 is not a hotspot residue for this antibody ( Figure 2), while for other antibodies, such as HCV1, N415 is a key binding site residue [8].
  • E2 434–446 bound to HC84.26AM is similar overall to the structure of these residues in the native E2 core protein, except for a significant deviation in C-terminal residues 443–446 that leads to a displacement of 7.7 ⁇ in the C ⁇ position of Lys446.
  • binding of HC84.26AM to the E2 core may induce conformational changes in residues 443–446 of the native protein.
  • N415P maintained wild-type levels binding across the panel of mAbs ( Figure 9), while H445P exceeded wild-type binding levels for the panel of mAbs ( Figure 11).
  • other mutants near domain D had little (A439P, N448Q) or negative effects (A440C/W616C) on mAb binding ( Figure 10), in the context of a genotype 1b E2 protein.
  • a set of mutants designed to insert glycans at specific sites in antigenic domain A were also produced. They were selected based on alanine scanning data (Figure 12), which provided the key antibody hotspot residues in this region.
  • the two crystal structures of E2 core contain this region, and were analyzed for residue surface exposure and potential to mutate residue pairs to N-glycan codons (NxT/NxS) without disrupting the E2 core fold ( Figure 13).
  • SEQ ID NO: 1 E2 polypeptide. H77 (genotype 1 a). aa 384-746 (NCBI refseq ID NP 751921 .1 )
  • Table 4 Computational analysis of domain D epitope conformation and proline substitutions, using the HC84-26AM-bound epitope structure (PDB code 4Z0X). Table 5. Isothermal calorimetry affinity data for E2 domain E peptide (aa 412-423) and proline mutants binding to the HC33.1 antibody.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • Communicable Diseases (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne des compositions et des méthodes se rapportant à la protéine E2 du VHC et des modifications de cette dernière qui améliorent l'immunogénicité de la protéine pour le développement de vaccins en termes de génération d'une réponse immunitaire neutralisante.
PCT/US2016/020720 2015-03-04 2016-03-03 Conception rationnelle de vaccins contre le virus de l'hépatite c WO2016141201A2 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562127911P 2015-03-04 2015-03-04
US62/127,911 2015-03-04
US201562216485P 2015-09-10 2015-09-10
US62/216,485 2015-09-10

Publications (2)

Publication Number Publication Date
WO2016141201A2 true WO2016141201A2 (fr) 2016-09-09
WO2016141201A3 WO2016141201A3 (fr) 2017-01-05

Family

ID=56848773

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/020720 WO2016141201A2 (fr) 2015-03-04 2016-03-03 Conception rationnelle de vaccins contre le virus de l'hépatite c

Country Status (1)

Country Link
WO (1) WO2016141201A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022104081A3 (fr) * 2020-11-12 2022-06-23 University Of Maryland, College Park Glycoprotéines e1e2 du virus de l'hépatite c (vhc) sécrétées modifiées et procédés d'utilisation associés

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6020122A (en) * 1995-06-07 2000-02-01 Abbott Laboratories Hepatitis C virus second envelope (HCV-E2) glycoprotein expression system
US20050053617A1 (en) * 2003-05-28 2005-03-10 Erik Depla Modified HCV NS3
EP2600894B1 (fr) * 2010-08-04 2017-02-01 The Macfarlane Burnet Institute For Medical Research And Public Health Ltd Protéines de virus de l'hépatite c modifiées

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022104081A3 (fr) * 2020-11-12 2022-06-23 University Of Maryland, College Park Glycoprotéines e1e2 du virus de l'hépatite c (vhc) sécrétées modifiées et procédés d'utilisation associés

Also Published As

Publication number Publication date
WO2016141201A3 (fr) 2017-01-05

Similar Documents

Publication Publication Date Title
US10759846B2 (en) Middle east respiratory syndrome coronavirus immunogens, antibodies, and their use
US11130785B2 (en) Prefusion RSV F proteins and their use
Vietheer et al. The core domain of hepatitis C virus glycoprotein E2 generates potent cross‐neutralizing antibodies in guinea pigs
JP2023533772A (ja) SARS-CoV-2及びインフルエンザ混合ワクチン
JP2023515800A (ja) コロナウイルス感染症2019(covid-19)の検出、予防及び治療のためのデザイナーペプチド及びタンパク質
ES2237115T3 (es) Particulas de proteinas de la envoltura del hcv: uso para la vacunacion.
JP2023524054A (ja) ベータコロナウイルスの予防と治療
US20150118242A1 (en) Hepatitis c virus neutralizing antibody
US20230338507A1 (en) Hiv-1 env fusion peptide immunogens and their use
AU2015313650B2 (en) Flavivirus virus like particle
JP2023524879A (ja) 安定化されたコロナウイルススパイクタンパク質融合タンパク質
US20230381293A1 (en) Stabilized norovirus virus-like particles as vaccine immunogens
EP3039036B1 (fr) Nouveaux antigènes et anticorps de la grippe
JP2015521592A (ja) 安定化されたgp120
KR101966841B1 (ko) 지카바이러스 e 단백질 유래의 재조합 항원 및 이의 용도
US9732121B2 (en) Rational vaccine design for hepatitis C virus
WO2016141201A2 (fr) Conception rationnelle de vaccins contre le virus de l'hépatite c
WO2017008049A1 (fr) Anticorps hétérotypiques spécifiques contre le rotavirus humain
US9751931B2 (en) Hepatitis C virus neutralizing antibodies and methods
US20240016923A1 (en) Modified secreted hepatitis c virus (hcv) e1e2 glycoproteins and methods of use thereof
WO2016115664A1 (fr) Anticorps monoclonaux anti-immunoglobuline a spécifiques des protéines 3d

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: 16759506

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16759506

Country of ref document: EP

Kind code of ref document: A2