MXPA00011508A - Antigenic complex comprising immunostimulatory peptide, cd4, and chemokine receptor domain for hiv treatment and immune disorders - Google Patents

Antigenic complex comprising immunostimulatory peptide, cd4, and chemokine receptor domain for hiv treatment and immune disorders

Info

Publication number
MXPA00011508A
MXPA00011508A MXPA/A/2000/011508A MXPA00011508A MXPA00011508A MX PA00011508 A MXPA00011508 A MX PA00011508A MX PA00011508 A MXPA00011508 A MX PA00011508A MX PA00011508 A MXPA00011508 A MX PA00011508A
Authority
MX
Mexico
Prior art keywords
peptide
sequence
ident
sec
antigen
Prior art date
Application number
MXPA/A/2000/011508A
Other languages
Spanish (es)
Inventor
Chang Yi Wang
Original Assignee
United Biomedical Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by United Biomedical Inc filed Critical United Biomedical Inc
Publication of MXPA00011508A publication Critical patent/MXPA00011508A/en

Links

Abstract

The invention provides peptides comprising a sequence homologous to a portion of the CDR-2 like domain of CD4, covalently linked to a helper T cell epitope, and optionally to other immunostimulatory sequences as well. The invention provides for the use of such peptides as immunogens to elicit the production in mammals of high titer polyclonal auto-antibodies, which are specific to CD4 surface complex. These auto-antibodies prevent binding of HIV viral particles to CD4+ cells. The peptides are useful in pharmaceutical compositions, to provide an immunotherapy for HIV infection and to protect against HIV infection.

Description

PEPTIDE COMPOSITION FOR PREVENTION AND TREATMENT PE HIV INFECTION AND DISEASES IMMUNES FIELD OF THE INVENTION This invention is directed to the use of a peptide composition as an immunogen, comprising each peptide contained therein, a target antigenic site that is recognized by antibodies against a host cell host / co-receptor complex for HIV. The CD4 complex associated with a chemokine receptor domain. The target antigenic site is in a cyclic form linked covalently, linearly and in tandem to (1) a carrier protein through chemical coupling, or preferably to (2) an attendant T cell epitope and other immunostitory peptide sequences by chemical coupling or more preferably by direct synthesis. More particularly, the present invention relates to the use of such a peptide composition as an immunogen to produce production in healthy mammals, including humans, of high titer antibodies having broad neutralized activities against primary isolates of all HIV isolates. Type 1 (HIV-1) and primary isolates of HIV type 2 (HIV-2). The present invention is also directed to a method for utilizing the peptide composition as an immunogen for the prevention and treatment of immunodeficiency virus infection as well as for the treatment of undesirable immune responses such as rejection to transplantation, and autoimmune disorders such as arthritis. rheumatoid, systemic lupus erythematosus, and psoriasis.
BACKGROUND OF THE INVENTION Despite intensive research for a vaccine in the 14 years since the discovery and characterization of HIV, major obstacles remain for the development of HIV vaccine and immunotherapy. These obstacles include HIV-1 variability, a lack of understanding of the structure of the virus, and a lack of understanding of the immune responses necessary for the prevention of HIV infection. See D. Burton and J. Moore, Nature Medicine, 1998, 4: 495-48. The chairman of the committee for the investigation of the AIDS vaccine of the government of the United States, declared on February 1, 1998 that a safe vaccine to prevent AIDS could be even further from a decade of testing, because much remains unknown about how the body's immune system works (http://cnn.com/HEALTH/ 9802/01 / aids. vaccine. search). There was early optimism for effective recombinant subunit HIV-1 envelope vaccines (eg, gp120 and gp160 vaccine products) since vaccine sera from various clinical trials were able to neutralize HIV-1 laboratory isolates in vitro (Belshe et al., J. Am. Med. Assoc, 1994, 272: 475; Keefer et al., AIDS Res. Hum Retroviruses, 1994, : 1713). This optimism was shocked when it was found that vaccine serum was very ineffective in neutralizing isolates from , V1H-1 of primary patients (Hanson, AIDS Res. Hum Retroviruses, 1994, 10: 645, Mascóla et al., J Infecí Dis., 1996, 173: 340). These disappointing discoveries led to NIH to decide in June 1994 to postpone costly large scale tests of various recombinant envelope proteins based on HIV subunit vaccines. HIV vaccine research now focuses on primary isolates which are believed to be more similar to HIV strains responsible for human infection than commonly used laboratory strains (Sawyer et al., J Virol, 1994, 68: 1342; Wrin e al., J Virol, 1995, 69:39). Primary isolates of HIV-1 are obtained by limited culture of plasma patient PBMCs with PBMCs without infecting. Primary viruses can be easily distinguished by phenotype as they were subsequently available from the adapted T-cell line (TCLA) viruses such as Iilb / LAI, SF2 and MN, which have passed over time in human T-lymphoid cell lines and have become well adapted to grow in these T cell lines: (1) Unlike TCLA viruses, most of primary isolates do not grow easily in T cell lines (2) Unlike TCLA viruses, which induce all syncytia, primary isolates include isolates that include syncytium (SI) that induce syncytium formation in PBMC culture and isolates that do not induce syncytium (NSI). Among the SI primary isolates, most will replicate especially in the MT2 line of HIV-sensitive T cells, but a few may replicate in the less permissible T-cell lines such as CEM or H9 that are commonly used for the culture of TCLA isolates . The primary isolates that do not induce syncytium (NSI) replicate only in primary T cells. (3) The primary isolates are highly resistant to in vitro neutralization by recombinant soluble forms of the viral CD4 receptor protein (rsCD4) which requires 200-2700 times more rsCD4 than the TCLA strains for comparable neutralization (Daar et al., PNAS USA, 1990, 87: 6574-6578). (4) Primary isolates are also resistant to neutralizing antibodies produced by the use of gp120 (envelope) vaccines. In contrast, TCLA strains are sensitive to neutralization by antibodies with specificities for the viral envelope (Sawyer et al., J Virol, 1994, 68: 1342; and, Mascóla et al., 1996). These phenotypic characteristics of primary isolates are due to poorly understood structural features of HIV, particularly the inaccessible quality of the viral envelope with respect to anti-env antibodies (D. Burton and J. Moore, Nature Medicine, 1998, 4: 495- 498). Viral variability, a genotypic characteristic, also remains an obstacle to the development of globally efficient HIV vaccines (Mascóla et al., 1996). These factors together have the unexpected failure of the virally directed AIDS vaccines that developed against TCLA homotypic strains already grown. An alternative proposal for the development of HIV vaccine could be through the intervention of HIV receptors in the host cell, thereby blocking the infection by preventing HIV from binding to, or binding with, susceptible cells. The cell-targeted proposal offers methods to overcome the hypervariability of the HIV envelope and phenotypic diversity. A proposal aimed at cells for the protection of HIV infection was suggested by the active and passive immunization studies in the rhesus SIV macaque model which showed that the anti-cell antibodies contributed greatly to the protection of the infection ( Stott, Nature, 1991, 353: 393). In addition, monoclonal antibodies directed against CD4, MHC class II molecules for the T cell receptor and the primary receptor for HIV binding have long been known to block infection in HIV-neutralization tests. 1 in a form that is dependent on the CD4 epitope, not the virus strain (Sattentau et al., Science, 1986; 234: 1120). In this proposal for immunoprophylaxis it has been found that anti-CD4 monoclonal antibodies are effective in blocking infection of cells by primary isolates (Daar et al., Proc. Natl. Acad. Sci. USA, 1990; 87: 6574; and Hasunuma e to al., J Immunol., 1992; 148: 1841). Other approaches directed to potentially effective cells include selecting chemokine receptors CXCR4, CCR5, CCR2b, and CCR3 that have recently been identified as co-receptors for HIV (Feng et al., Science, 1996; 272: 872; and, Doranz et al. , Cell, 1996; 85: 1149). These co-receptors work together with CD4 to initiate post-link interaction of the glycoprotein viral envelope with the host cell membrane and in post-entry steps of retrovirus replication (Chackerian et al., J Virol, 1997; 71: 3932). The requirement for efficient binding and fusion of HIV for CD4 and a co-receptor suggests that either or both of these molecules may be good targets for strategies directed at cells to inhibit infection. Antibodies directed to a CD4 host cell / co-receptor complex have been shown to affect the binding and post-binding stages of HIV infection (Wang, WO 97/46697). These antibodies neutralized the transmission of virus to cell or cell to cell of HIV strains that induce (SI) and that do not induce (NSI) syncytium. A chemokine antagonist that binds to CCR5 has also been shown to be effective in preventing infection by SI and NSI viruses (Simmons et al., Science, 1997; 276: 276). The neutralization of NSI isolates is particularly significant since NSI strains are believed to be responsible for most HIV transmission and are frequently resistant to anti-HIV antibodies that neutralize TCLA isolates (Fauci, 1996). The agents that select HIV cellular receptors avoid the need to confront diverse phenotypes and the hypervariability of the viral envelope and also offer potential neutralization activity against HIV-2 and SIV, (Chen et al, J Virol, 1997; : 2705; Pleskoff et al., J Virol, 1997; 71: 3259; WO 97/46697). A receptor / co-receptor host cell complex comprising CD4 and a chemokine co-receptor on the surface of the T host cells, which facilitates viral binding and entry into the host T cells, is reported to be an effective target for neutralizing antibodies in a co-pending patent application (WO 97/46697). In this application, the current inventor demonstrated that the antibodies raised are specifically directed against this complex cellular surface antigen. No other anti-cell antibody arises in response to cell surface antigens in the HPB-ALL cells of neutralized HIV-1 primary isolates. Antibodies with the desired properties as described in this application can block infection of monkeys by SIV, HIV-1 immune system infection in vivo reconstituted in mice, in vitro infections of human cells by HIV-1 primary isolates in human cells by HIV-2 This complex cell surface antigen comprising the CD4 receptor associated with a chemokine co-receptor (CD4 / co-receptor complex) acts as a target for anti-cell protective antibodies. Anti-cell antibodies to the CD4 / co-receptor complex present a more effective pattern of neutralization against HIV-relevant strains than anti-virus antibodies directed against the viral envelope. As shown in the co-pending application (WO 97/46697), a monoclonal antibody (MAb B4), produced against HPB-ALL, reacts moderately against recombinant soluble CD4 protein (rsCD4) and binds strongly to HPB cells -ALL. The specificity for the CD4 cell surface complex / co-receptor was found to be highly effective in neutralizing primary isolates of HIV-1, but less effective in neutralizing strains of TCLA. In contrast, anti-env antibodies present an inverse pattern for the preferential neutralization of TCLA strains. It was found that MAb B4 neutralized primary isolates of HIV in an in vitro microplate test at a concentration of < 10 μg / ml. In contrast, polyclonal antibodies with high titer (> 5 Log10) of specificity for recombinant soluble CD4 (rsCD4) failed to present any neutralizing activity for primary HIV isolates despite their strongly T cell-binding activities. Thus, isolates Primary antibodies appear to be preferentially sensitive to antibodies with specificity for the CD4 cell surface antigen / co-receptor complex, as compared to antibodies with a pure CD4 specificity. Extensive characterization of HIV neutralization by antibodies to the anti-CD4 / co-receptor complex includes MAb B4 and its homologs MAb M2 and MAb B13 (WO 97/46697). The mechanism for the broad neutralizing activity of antibodies to the CD4 / co-receptor complex is unclear due to the diverse roles of this cell surface complex in mediating HIV infection, as shown by the ability of these antibodies to affect the linkage and post-linkage stages of HIV infection (Wang, WO 97/46697). The CD4 cell surface complex / co-receptor can play dual roles in mediating HIV infection and pathogenesis: (1) as a T cell surface receptor for HIV binding, fusion and entry into HIV cells; or (2) as a factor that suppresses HIV. However, even when these agents are effective for the inhibition of HIV infection, antagonists or antibodies directed to previous cells can not be used as preventive vaccines. Antagonists or antibodies directed to neutralizing (Burkly et al., J Immunol, 1992; 149: 1779) and the widely neutralizing anti-CD4 / co-receptor monoclonal antibody reported by Wang (WO 97/46697) recognizes discontinuous conformational sites on CD4 that can not be easily duplicated. Without accurate knowledge of the vulnerable sites, the selection of the extended recombinant immunogens useful as antigenic targets of host cells and the reproduction thereof is very difficult. Most antibodies raised by immunization with CD4 lack useful specificities (Davis et al., Nature, 1992; 358: 76). For example, the high titer hyperimmune antiserum rsCD4 was devoid of neutralizing activity for primary isolates of HIV (WO 97/46697). In addition, antibodies with broad reactivity for extensive regions of a T cell antigen are expected to be highly immunosuppressive (Reimann et al., AIDS Res. Hum Retroviruses, 1997; 13: 933). In addition, although extensive CD4 mapping studies have produced a functional structure map for the molecule (Sattentau et al., Science, 1986, 234: 1120; Peterson and Seed, Cell, 1988, 54:65; Jameson et al. ., Science, 1988, 240: 1335, Sattentau et al., J Exp. Med., 1989, 170: 1319, Hasunuma et al., J Immunol, 1992, 148: 1841, Burkly et al., J Immunol, 1992. , 149: 1779; Davis et al., Nature, 1992, 358: 76), this mapping does not provide structural models of sufficient precision to predict vulnerable effector sites that can be duplicated as synthetic peptides. The availability of models for CD4 does not describe useful immunogens based on CD4. In addition to the appropriate site specificity, the immunogenic receptor / co-receptor of an effective HIV vaccine must be highly immunostimulatory to evoke antibody responses of sufficient level of protection. These immunogens must also be designed to overcome the strong tolerance presented to the own molecules. Thus, a need also remains for immunogens of sufficient immunopotency. It is an object of the present invention to provide peptide compositions having the desired site specificity and immunopotency, as immunogens for the prevention of HIV infection. The improved immunogenicity and specificity for the useful synthetic peptide immunogens of the present invention have been achieved through the incorporation of a collection of methods for the identification and design of synthetic peptide immunogens. These methods include: (1) an effective method for the identification of an effective epitope of high target affinity; (2) means for the stabilization of the conformational characteristics of this target site on a synthetic peptide by the introduction of cyclic repressions, to maximize the cross-reactivity of the native molecule; (3) means for increasing the immunogenicity of the B-cell target epitope by combining it with a site comprising a highly reactive promiscuous promiscuous T (Th) helper cell epitope; and (4) the means for enlarging the repertoire of T-cell epitopes by application of combinatorial peptide chemistry and thereby further accommodating the immuno-variable responsiveness of a population produced by mixtures of races. Synthetic peptides have been used for "epitope mapping" to identify immunodominant determinants or epitopes on protein surfaces for the development of new vaccines and diagnostics. The epitope mapping employs a series of overlapping peptides corresponding to regions on the protein of interest to identify sites that participate in the antibody-immunogenic determinant interaction. Commonly, epitope mapping employs peptides of relatively short length to detect precisely linear determinants. A rapid method of epitope mapping known as PEPSCAN is based on the simultaneous synthesis of hundreds of overlapping peptides, of lengths of 8 to 14 amino acids, coupled to solid supports. Coupled peptides are tested for their ability to bind antibodies. The PEPSCAN proposal is effective for locating linear continuous determinants, but not for the identification of epitopes that are needed for the imitation of discontinuous effector sites such as the HIV receptor / co-receptor binding site (Meloen et al., Ann Biol. Clin. ., 1991; 49: 231-242. An alternative method lies in a set of overlapping and nesting peptides of multiple lengths ranging from 15 to 60 residues. These longer peptides can be synthesized reliably, but laboriously by a series of independent solid phase peptide synthesis, rather than by rapid and simultaneous PEPSCAN syntheses. The resulting set of overlapping and nesting peptides can then be used for analysis of antibody binding in systems such as experimental immunizations and natural infections, to identify long peptides having better immunodominant determinants, including simple discontinuous epitopes. This method is exemplified in the Wang studies for the mapping of immunodominant sites of HTLV l / ll (US 5, 476, 765) and HCV (US 5, 106, 726). It was used for the selection of a precise position on the gp120 sequence for optimal presentation of an HIV neutralizing epitope (Wang et al., Science, 1991; 254: 285-288). Peptide immunogens are generally more flexible than proteins and do not tend to retain any preferred structure. Therefore, it is useful to stabilize a peptide immunogen by the introduction of cyclic repressions. A properly cyclized peptide immunogen can mimic and preserve the conformation of a selected epitope and thereby evoke antibodies with cross reactivities for that site on the authentic molecule. For example, a wavy structure present on an authentic epitope can be duplicated more precisely on a synthetic peptide by the addition of cysteine residues advantageously placed followed by cyclization through sulfhydryl groups (Moore, Chapter 2 in Synthetic Peptides: A User's Guide, ed. Grant, WH Freeman and Company: New York, 1992, pp. 63-67). Another important factor affecting the immunogenicity of a peptide immunogen derived from a receptor / co-receptor complex is the presentation of this peptide to the immune system by epitopes of T-helper cells that react with a host cell receptor T and Class II molecules. MHC (Babbitt et al., Nature, 1985; 317: 359-361). The attendant T (Th) epitopes are frequently provided by carrier proteins with concomitant disadvantages due to difficulties in making well-defined peptide carrier conjugates, misdirection of most of the antibody response to the carrier, and carrier-induced epitope deletion (Cease). , Intern Rev Immunol, 1990; 7: 85-107; Schutze et al., J Immunol, 1985; 135: 2319-2322). Alternatively, the T cell support can be stimulated by synthetic peptides comprising the Th sites. Thus, Class II Th epitopes, called promiscuous Th evoke sufficient T cell support and can be combined with B cell epitopes that are poor in themselves. immunogenic to generate potent peptide immunogens (US 5, 759, 551). Well-designed promiscuous Thi-B epitope chimeric peptides are capable of producing Th responses and resulting antibody responses selected for B-cell sites in most members of a genetically diverse population expressing various MHC haplotypes. Promiscuous Th may be provided by specific sequences derived from potent foreign antigens, such as, for example, measles virus F protein, hepatitis B virus surface antigen, and Chlamydia trachomatis major end membrane protein (MOMP). Many known promiscuous Th have been shown to be effective in enhancing a poorly immunogenic peptide corresponding to the decapeptide hormone (US Patent No. 5, 759, 551). Promiscuous Th epitopes vary in size from about 15 to about 40 amino acid residues in length (US Patent 5, 759, 551), and frequently share common structural characteristics and may contain specific tag sequences. For example, a common feature are antipathetic helices, which are alpha-helical structures with hydrophobic amino acid residues that dominate one side of the helix and with charged and polar residues that dominate the surrounding faces (Cease et al., Proc Natl Acad Sci USA, 1987; 84: 4249-4253). Th epitopes frequently contain additional primary amino acid patterns such as a charged or Gly residues followed by two or three hydrophobic residues, followed in turn by a charged or polar residue. This pattern defines what is called Rothbard sequences. Also, Th epitopes frequently obey the rule of 1, 4, 5, 8 where a positively charged residue is followed by hydrophobic residues in the fourth, fifth and eighth positions, after the charged residues, consisting of an antipathetic helix that has the positions 1, 4, 5, and 8 located on the same side. Since all these structures are composed of commonly hydrophobic charged and polar amino acids, each structure can exist simultaneously within a single Th epitope (Partidos et al., J Gen Virol, 1991; 72: 1293-99; Alexander et al., Immunity. , 1994; 1: 751-761). Most, if not all, of the promiscuous T-cell epitopes contain at least one of the periodicities described above. These characteristics can be incorporated into the designs of "idealized artificial Th sites". Promiscuous Th epitopes derived from foreign pathogens include, but are not limited to, T-cell epitopes of core antigen and hepatitis B surface (HBS Th and HBC Th), T-cell epitopes of pertussis toxin assistants (PT Th), tetanus toxin assistant T cell epitopes (TT Th), T cell epitopes, F protein assistants of measles virus (MVF Th), epitomes of T-cell assistants of the outer main membrane of Chlamydia trachomatis ( CT Th), diphtheria toxin attendant T cell epitopes (DT Th), attendant T cell epitopes of Plasmodium falciparum circumsporozoite (PF Th), epitopes of Trisos phosphate isomerase isomerase T cells from Schistosoma mansoni (SM Th), and T-cell epitopes of Trat Escherichia coli (Trat Th). Th derived from pathogen was listed as SEC. FROM IDENT. US Nos. 2-9 and 42-52 in US Patent 5,759,551; as a P11 Chlamydia assistant site in Stagg et al., Immunology, 1993; 79; 1-9; and as peptide HBc 50-69 in Ferrari et al., J Clin Invest, 1991; 88: 214-222. Useful Th sites may also include combinatorial Th that incorporate selected degenerate sites within the design of the idealized Th sites. In Wang ei al. (WO 95/11998), a particular class of a combinatorial epitope was designed as a "Structured Synthetic Antigen Library" or SSAL. A Th constructed as an SSAL epitope is composed of location substitutions organized around a structural framework of unchanged residuals. The sequence of the SSAL is determined by aligning the primary amino acid sequence of a promiscuous Th, which retains residues relatively unchanged at the positions responsible for the unique structure of the Th peptide and which provides degeneracy at the positions associated with recognition of the various MHC restriction elements. Lists of variable and invariant positions and preferred amino acids are available for MHC-binding motifs. (Meister et al., Vaccine, 1995; 13: 581-591; Alexander et al., Immunity, 1994; 1: 751-761). All members of the SSAL are produced simultaneously in a simple solid phase peptide synthesis in tandem with the B cell epitope selected from other sequences. The sequence of the Th library maintains the structural motifs of a promiscuous Th and accommodates reactivity in a wider range of haplotypes. For example, the degenerated Th epitope described as SSAL1TH1 was modeled after a promiscuous epitope was taken from the measles virus F protein (Partidos et al., 1991). SSAL1TH1 was used in tandem with an LHRH target peptide. Like the epitope of measles, SSAL1TH1 follows the sequence of Rothbard and the rule of 1, 4, 5, 8: 1 51015 Asp-Leu-Ser-Asp-Leu-Lys-Gly-Leu-Leu-Leu-His-Lys- Leu-Asp- Gly-Leu Glu lie Glu lie Arg lie Me Arg Me Glu lie Val Val Val Val Val Val Phe Phe Phe Phe Phe Phe Phe The residues loaded with Glu or Asp are added in position 1 to increase the load which surrounds the hydrophobic face of the Th. The hydrophobic face of the antipathetic helix is then maintained by the hydrophobic residues in the hydrophobic., 5, 8, 9, 10, 13, and 16 with variability in 2, 5, 8, 9, 10, 13 and 16 to provide a facade with the ability to link to a wide range of MHC constraint elements. The net effect of the SSAL characteristic is to increase the range of immunoresponsibility to an artificial Th (WO 95/11998).
In accordance with the present invention, the peptide immunogens that are effective for HIV have been designed with precise epitope mapping, cyclic repressions, the incorporation of promiscuous or promiscuous Th epitopic Th and idealized SSAL epitopes. Such peptides are preferred because they are safe and effective. It is believed that the peptide immunogens of the present invention provide immunopotence due to the effective presentation of an HIV receptor / co-receptor binding site which is optimized through precise optimization and cyclization and the use of Th responsive epitopes widely. reagents BRIEF DESCRIPTION OF THE INVENTION The peptide compositions of the present invention comprise one or more peptide immunogens that have been designed as discussed above. Peptide compositions are the basis for a vaccine for the prevention and effective treatment of HIV infection and immune disorders. The component peptides of the invention are preferred by their presentation to neutralize receptor / co-receptor effector sites of the CDR2-like domain of CD4. These peptides evoke effective antibody responses by (1) having optimized site specificity, obtained by accurate epitope mapping of the CDR2-like domain and selective cyclization taking into consideration the native conformation of CDR2 and by (2) its Th responsivity broadly. reactive In accordance with the present invention, one or more peptides comprising each of the peptides have been provided with one of two peptide sequences corresponding to the effector sites located in the CDR2 domain of CD4, or immunologically functional analogues thereof. In addition, the target sites of the peptides of the invention become more immunogenic by means of covalent linkage to a carrier protein through chemical coupling, or more preferably by means of covalent linkage to synthetic immunostimulatory elements (such as promiscuous Th epitopes). , through chemical coupling or more preferably through direct peptide synthesis. Specific examples of carrier protein and immunostimulatory elements are provided, for example, lamellated hemocyanin carrier (KLH), modified pertussis enterotoxin A (PEA), Th epitopes (eg, SEQ ID NO: 6), and general immunostimulatory peptides (e.g., the invasin peptide (Inv) from Yersinia (SEQ ID NO: 7)). The synthetic peptides of the invention can be represented by the formulas: (A) n- (Th) m- (B) 0- (CD4-CDR2 peptide antigen) -X or (A) n- (CD4-CDR2 peptide antigen) - (B) 0- (Th) mX (CD4-CDR2 peptide antigen) - (B) 0- (Th) m- (A) nX or (Th) m- (B) 0- (CD4-CDR2 peptide antigen) - (A) nX wherein: each A is independently an amino acid, or a general immunostimulatory peptide; each B is independently an amino acid or other chemical bond; X is an amino acid a-COOH or a-COHN2; Th is an attendant T-cell epitope or a homologue that enhances immunity or the segment thereof; "CD4-CDR2 antigen peptide" is a peptide antigen that evokes antibodies that react with the CD4 surface complex; n is from 1 to about 10; m is from 1 to about 4; I is from 0 to about 10. The peptide compositions of the present invention comprise peptide immunogens of from about 30 to about 115 amino acid residues, preferably from about 40 to about 90 amino acid residues and more preferably from about 50 to about 80 residues of amino acids.
The compositions of the present invention additionally further comprise adjuvants and / or delivery vehicles and other ingredients routinely incorporated with the vaccine formulations. The present invention provides instructions for dosing such that immunotherapeutic antibodies directed against the selected CD4-CDR2 effector sites are generated. The present invention provides, for the first time, synthetic peptides capable of producing antibodies in mammals that are protective against infection by primary isolates of HIV of multiple ciados. The antibody response to the peptide compositions of the invention provides protection or therapy against HIV infection of a host by: (1) blocking the binding of HIV to cells expressing CD4, (2) blocking the formation of induced syncytium for HIV among CD4-expressing cells, (3) effectively neutralizing in vitro infection of CD4 positive cells by primary isolates of all HIV-1 and HIV-2 types, and (4) preventing infection by primary HIV isolates; when a vaccine formation comprising a peptide composition of the present invention is administered to the host. Peptide compositions are useful for the prevention and treatment of HIV infection by primary isolates of all HIV-1 isolates and primary isolates of HIV-2 as well as for the treatment of immune responses mediated by undesirable CD4 cells, such as rejection. of transplantation, and autoimmune disorders, such as rheumatoid arthritis, systemic lupus erythematosus and psoriasis.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 represents the amino acid sequence of CD4 (SEQ ID NO: 1) human, a part of the complex CD4 / co-receptor host cell antigen as deduced from the nucleic acid sequence. The amino acids are represented by the standard simple letter codes as follows: Wing: A Cys: C His: H Met: M Thr: T Arg: R Gln: Q lie: I Phe: F Trp: W Asn: N Glu: E Leu: L Pro: P Tyr: Y Asp: D Gly: G Lys: K Ser: S Val: V The numbering system is that of Littman et al. . { Cell, 1988, 55: 541. The underlined region (AA27-AA66) is the region from which the CD4-CDR2 antigen peptides of the invention are derived.
DETAILED DESCRIPTION OF THE INVENTION As used herein, "primary isolates of human immunodeficiency virus type 1 (HIV-1)" are obtained by limited culture, up to five passages, on peripheral blood mononuclear cells (PBMC) from donors. The primary isolates can be distinguished from laboratory strains adapted to T cell lines (TCLA) such as lllb / LAI, SF2 and MN that have been passed down over time in human T lymphoid cell lines. First, the most primary isolates do not grow easily in T cell lines and have phenotypes that induce (SI) and do not induce syncytium (NSI). For example, many SI primary isolates that induce syncytium formation in PBMC cultures will replicate in especially HIV sensitive MT2 T cell lines, but a few will replicate in less permissive T cell lines such as CEM or H9. Primary NSI isolates will replicate only in primary T cells. Second, they differ from the TCLA strains in their sensitivity to in vitro neutralization by soluble recombinant forms of the CD4 viral receptor protein (rsCD4) (Daar et al., PNAS USA, 1990, 87: 6574-6578). Third, the strains adapted to the laboratory are sensitive to neutralization by antibodies with specificities for the viral envelope, while the primary isolates are resistant (Sawyer et al., J Virol, 1994, 68: 1342; Mascóla et al., J Infecí Dis, 1996, 173: 340). As used herein, "CD4" means any CD4 protein encoded by a naturally occurring CD4 gene. CD4 was initially described as a cell surface marker for T helper lymphocytes. CD4 was subsequently found to be poorly expressed on monocytes, Langerhans, microglial cells, and subsets of B cells. It was also found that the CD4 molecule participates directly in the activation of T-helper cells specific for antigen through its function as a receptor for the MHC class II molecule. In 1984, it was found that human CD4 is the receptor for HIV (Dalgleish et al., Nature, 1984, 312: 763). The binding of HIV envelope glycoprotein, gp120 to CD4 represents the initial stage of viral entry into the target cell. The amino acid sequence for human CD4 is incorporated herein by Maddon et al. . { Cell, 1985; 42:93; and, Littman et al., Cell, 1988; 55: 541) and shown as Figure 1 and SEC. FROM IDENT. NO: 1. As used herein, "recombinant soluble CD4" or "rsCD4" is a polypeptide expressed by recombinant microorganisms in cells consisting of AA AA375 of human CD4 (Figure 1, SEQ ID NO: 1) . As used herein, "surface CD4 complex" or "surface complex comprising CD4" or "receptor / co-receptor complex comprising CD4" refers to the intact native CD4 protein, as it appears in its natural context on the surface of mammalian cells, together with and / or complex with any associated membrane protein. As used herein, the term "immunogen" refers to a peptide composition, which, when administered to a host, is capable of inducing antibodies against target effector sites present on the CDR2 domain of CD4 (SEQ. DE IDENT nos: 2 and 3), leading to high titre antibodies that have broad neutralizing activities against primary isolates of all types of HIV type 1 (HIV-1) and type 2 (HIV-2). The target CDR2-CD4 sites are underlined in Figure 1 and listed as SEC. FROM IDENT. NOS: 2 and 3. A "CD4-CDR2 antigen peptide" according to the present invention, is between about 25 and about 50, preferably between about 30 and about 46, amino acids in length, and contains two cysteine residues separated by an interposed sequence of 28 to 40 amino acid residues. The intervening sequence can be any contiguous portion of the sequence represented by residues 27 to 66 of SEC. FROM IDENT. NO: 1, or it may be an immunologically functional homologue of residues 27 to 66 of SEC. FROM IDENT. NO: 1. A conjugated peptide, as used herein, refers to a molecule comprising a CD4-CDR2 antigen peptide covalently linked to an assistant epitope peptide Th, by any means to direct peptide synthesis of the molecule. Examples of the covalent coupling of a CD4-CDR2 antigen peptide with a Th epitope peptide to form a peptide conjugate is thiol-haloacetamide coupling, thiol-maleimide coupling, disulfide bonding, interchain thiol-thiol, and the like. A "peptide immunogen" as used herein refers to a peptide or conjugated peptide, comprising a CD4-CDR2 antigen peptide covalently linked to a Th epitope peptide, optionally additionally comprising a general immunostimulatory peptide, a linker , and a spacer as further described herein; and that it has the ability to evoke antibodies for the CD4-CDR2 antigen peptide. The term "homologous" as used herein refers to a peptide having essentially the same amino acid sequence, with conservative substitutions of up to about 10% of the amino acids. Conservative substitutions are those in which one amino acid is replaced by another, preferably of the same class (eg, hydrophobic, polar, charged, etc.), without significantly altering the properties of the peptide. Homologs can also have amino acid insertions or deletions that do not significantly alter the immunological properties of the peptide. The homologs can be obtained artificially, or they can be found as naturally occurring variants of the peptide sequences presented herein. Immunologically functional homologs are homologues that induce essentially the same reaction of the immune system, for example, responsiveness of T cells, responsiveness of B cells or induction of antibodies against a given antigen. This invention is directed to the use of novel peptide compositions as immunogens. The immunogens are useful for the generation, by active immunization, of high titer antibodies directed against the effector sites (SEQ ID NOS: 2 and 3) on the CDR2 domain of CD4 in mammals including humans. The immunogens of the present invention are useful for the prevention and treatment of infection of immunodeficiency viruses as well as for the treatment of immune responses mediated by undesirable CD4 + cells, such as transplant rejection and autoimmune disorders such as rheumatoid arthritis, systemic lupus erythematosus and psoriasis. Such interventions employed in the prevention and treatment of HIV infection and immune disorders through the use of specific CD4-reactive antibodies, i.e., a type of immunotherapy, can be achieved passively, through prophylactic treatment with "site-directed" antibodies. "specific, to a site on the CDR2-like domain of CD4. More preferably, as described herein, therapy can be effected through active immunization, by inoculating the host with a composition comprising one or more peptide immunogens of the present invention. These immunogens produce the production by the host of their own CD4-CDR2 reactive antibodies directed to the site, which have broad neutralizing activities against primary isolates, all of which are co-infected with HIV type 1 (HIV-1) and type 2 (HIV- 2). It is believed that active immunization will provide a more effective and longer-lasting form of protection than passive immunization.
The target sites of the CD4-like domain of CD4 (SEQ ID NOS: 2 and 3) are conformationally restricted by cyclization through the addition of cysteine residues to the N and C terminals (SEQ ID NOS: 4 and 5). Such target sites may also include immunological counterparts of SEC. FROM IDENT. NOS: 4 and 5 comprising 1-5 additional amino acids taken from any term of SEC. FROM IDENT. NOS: 2 and 3, with the proviso that the corrugated structure of the single disulfide is retained (for example, SEQ ID NOS: 10 and 11). The target sites are further modified within the immunogenic CD4-CDR2 antigen peptides by chemical coupling to a carrier protein, for example, slotted limpet hemocyanin (KLH) and modified pertussis enterotoxin A (PEA). A deficiency of such vaccines based on "CD4-CDR2 antigen peptide carrier protein" are (1) the weak immunogenicities of the target antigenic sites, an inherent problem associated with almost all of the antigens themselves; (2) the large portion of the non-functional antibodies directed against the carrier proteins and (3) the epitope suppression potential induced by the carrier. It is therefore preferable to render the immunogenic peptides by the tandem addition of chemically defined promiscuous Th and / or other immunostimulatory peptides, through chemical coupling or preferably through direct peptide synthesis. The preferred immunogens of the present invention minimize the generation of irrelevant antibodies to produce a more focused immune response to the "target sequences". The desired antibodies have reactivity to the CD4 surface complex, without producing undesirable side effects that may complicate the immunotherapy process for the prevention and treatment of HIV infection and immune disorders. In addition, antibodies specific for sites selected for the desired sites can be generated more widely in a genetically diverse host population by the use of promiscuous Th. These antibody responses lead to high titre antibodies having broad neutralizing activities against primary isolates of all types of HIV type 1 and type 2. The present invention is also directed to a method for using peptide compositions as immunogens for the prevention and treatment of infection of the immunodeficiency virus as well as for the treatment of immune responses mediated by undesirable CD4 cells, such as rejection to transplantation, and autoimmune disorders such as rheumatoid arthritis, systemic lupus erythematosus and psoriasis. Antibodies that are specific for the receptor / co-receptor complex on the surface of the host cell that comprises CD4, as distinguished from specific antibodies for which rsCD4, probably interact with the immune system in various ways: 1. They can block Class II CD4 interaction between T cells expressing CD4 and other activated T cells, B cells, or monocytes; 2. They can supply signals to the T cells, thus inhibiting the immunoregulatory functions mediated by normal CD4 + T cells; 3. They can induce cell death of cells expressing CD4 by apoptosis when triggered by a simultaneous binding of the T cell receptor molecules; and 4. Block the interactions between CD4 and HIV, which inhibits HIV-mediated immunopathology. Antibodies to the surface complex comprising CD4 are good candidates for preventing and treating HIV infection and diseases associated with HIV including AIDS. At a more general level, antibodies to the surface CD4 complex may be useful for preventing or curing undesirable immune responses mediated by CD4 expressing T cells, such as rejection of transplants, and autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus. , or psoriasis.
The properties of the antibodies generated by the peptide compositions of the present invention are summarized here based on the results obtained in Examples 1-5: 1. Link to rsCD4 in an ELISA test; 2. Link to cells expressing CD4 in an immunofluorescent test; and 3. Primary isolates of HIV resistant to neutralizing neutralization in an in vitro microplate test. Antibodies with these characteristics are especially useful for the prevention and treatment in humans of diseases caused by infectious agents whose primary targets are CD4-positive cells. Accordingly, the present invention provides peptide compositions as immunogens, useful for the prevention and treatment of diseases in humans caused by infectious agents whose primary targets are CD4 positive cells, particularly HIV-related diseases, which include all stages of AIDS. The present invention also provides methods for using these antibody compositions. The peptide compositions of the present invention comprise peptide immunogens that incorporate any of two peptide sequences that correspond to target effector sites located on the CDR2-like domain of CD4 (SEQ ID NOS: 2 and 3), or immunologically functional homologs of the same. The immunogens are characterized by their evocation of neutralizing antibodies against CD4 effector sites / co-receptor CD4 CD4 domain. The immunogens evoke protective antibody responses by virtue of their optimized site specificity, obtained by means of (1) accurate epitope mapping, (2) cyclization to repress their conformations in consideration of their native conformation; and (3) its response Th widely reactive. Specifically, the target sites are taken from the similar domain CDR2 of the native human CD4 sequence. The amino acid sequence for CD4 is incorporated herein by Maddon et al. . { Cell, 1985; 42:93; and, Littman et al., Cell, 1988; 55: 541) and shown as Figure 1 and SEC. FROM IDENT. NO: 1. The CD4-CDR2 target sites are shown underlined in Figure 1 and listed as SEC. FROM IDENT. NOS: 2 and 3. The peptide compositions of the present invention are preferably produced as synthetic peptides comprising the target sites (SEQ ID NOS: 2 and 3), in which the targets have been modified from their native sequences by the insertion of cysteine residues at or near the N-terminus and C-terminus to facilitate the formation of cyclic peptides (eg, SEQ ID NOS: 4 and 5). The peptide compositions of the invention also comprise immunological homologs of SEC. FROM IDENT. NOS: 4 and 5 which may comprise of 1-5 additional amino acids taken from any SEC terminal. FROM IDENT. NOS: 2 and 3 (for example, SEQ ID NOS: 10 and 11), with the proviso that the undulated simple disulfide structure is retained. The target site can also include immunologically functional homologs comprising a cyclic peptide in the range of about 25 to about 50 amino acids, which have an contiguous amino acid sequence derived from SEC. FROM IDENT. NOS: 2 and 3. The cyclic structure is an essential element of the invention, since the peptides comprising the linear counterparts of the target sites do not produce antibodies with neutralizing activity against the primary isolates of HIV. In addition, the target site of the peptides of the invention becomes immunogenic by means of the covalent linkage to a carrier protein to synthetic immunostimulatory elements, such as, for example, promiscuous Th epitopes derived from viruses and pathogenic bacteria, artificial promiscuous Th epitopes, and general immunostimulatory peptides. Specific examples of carrier proteins and immunostimulatory elements are provided, for example, slotted limpet haemocyanin carrier protein (KLH), modified pertussis enterotoxin A (PEA) carrier protein, hepatitis B virus surface antigen Th (SEQ ID NO: 8), an artificial Th ( for example, SEQ ID NO: 6), and a general immunostimulatory invasin peptide (Inv) from Yersinia (SEQ ID NO: 7). ~ The fully synthetic peptides of the invention can be represented by the formulas: (A) n- (Th) m- (B) 0- (CD4-CDR2 peptide antigen) -X or (A) n (CD4-CDR2 peptide antigen) - (B) 0- (Th) mX or (CD4- CDR2 peptide antigen) - (B) 0- (Th) m- (A) nX or (Th) m- (B) 0- (CD4-CDR2 peptide antigen) - (A) nX where: each A is independently a amino acid, a-NH2, or a general immunostimulatory peptide; each B is independently chosen from the group consisting of amino acids, -NHCH (X) CH2SCH2CO-, -NHCH (X) CH2SCH2CO (e-N) Lys-, -NHCH (X) CH2S-succinimidyl (e- N) Lys-, and -NHCH (X) CH2S- (succinimidyl) -; X is an amino acid a-COOH or a-CONH2; Th is an attendant T-cell epitope or immunomodulatory homologue or a segment thereof; "CD4-CDR2 antigen peptide" is as defined above, and is preferably SEC. FROM IDENT. NO: 4 or SEC. FROM IDENT. NO: 5, or a cross-reactive and immunologically functional homolog thereof; n is from 1 to about 10; m is from 1 to about 4; I is from 0 to about 10. The peptide compositions of the present invention comprise peptide immunogens of from about 30 to about 115 amino acid residues, preferably from about 40 to about 90 amino acid residues, and more preferably from about 50 to about 80 amino acid residues. When A is an amino acid, it can be any amino acid that occurs naturally or does not occur naturally.
Amino acids that do not occur naturally include, but are not limited to D-amino acids, β-alanine, ornithine, norleucine, norvaline hydroxypropylin, thyroxine, α-aminobutyric acid, homoserine, citrulline and the like. In addition, when m is larger than one, two or more of the groups A are amino acids, then each amino acid can be independently the same or different. When A is an invasin domain, it can be an immunostimulatory epitope of the invasin protein of a Yersinia species. This immunostimulatory property results from the ability of the invasin domain to interact with the ß1 integrin molecules present in T cells, particularly immunoactivated by memory T cells. The specific sequence found for an invasin domain that interacts with β1 integrins has been described by Brett et al. { Eur J Immunol, 1993; 23: 1608). A preferred embodiment of the invasin domain (Inv) for binding to a promiscuous Th epitope has previously been described in U.S. Patent 5,759,551, which is incorporated herein by reference. The Inv domain preferably has the sequence: Thr-Ala-Lys-Ser-Lys-Lys-Phe-Pro-Ser-Tyr-T r-Ala-Trir-Tyr-Gln-Phe (SEQ ID NO: 7) or is an immunostimulatory homologue thereof of the corresponding region in another invasin protein of Yersinia species. Such homologs may also contain substitutions, deletions or insertions of amino acid residues to accommodate variation from strain to strain, with the proviso that homologs retain immunostimulatory properties. The invasin domain is preferably linked through a spacer, provided by additional amino acids "A", to the Th peptide. In a preferred embodiment, n is 3 and (A) 3 is an invasin domain (Inv), glycine and glycine in this order. (B) 0 is an optional spacer and comprises amino acids which may occur naturally or amino acids that do not occur naturally as described above. Each B is independently the same or different. The carrier proteins are covalently linked to the peptides with a spacer (eg, Lys-Lys-Lys) by chemical coupling. The amino acids of (B) 0 may also provide a spacer, for example, Gly-Gly or eNLys, between the promiscuous Th epitope and the CD4-CDR2 antigen peptide (SEQ ID NOS: 4 and 5), to evoke sufficient antibody responses. In addition to physically separating the Th epitope from the B cell epitope (e.g., SEQ ID NOS: 4 and 5) and immunological homologs thereof, a spacer such as Gly-Gly can disrupt any secondary artifact structure created by the binding of the Th epitope to the CDR-CDR2 antigen peptides and thereby eliminating the interference between the Th and / or B cell responses. The amino acids of (B) 0 can also form a spacer which acts as a flexible hinge that improves separation of Th and IgE domains. Examples of the sequences encoding flexible hinges are found in the immunoglobulin heavy chain hinge region. The flexible hinge sequences are frequently rich in proline. A particularly useful flexible hinge is provided by the sequence Pro-Pro-Xaa-Pro-Xaa-Pro (SEQ ID NO: 9), wherein Xaa is any amino acid, and preferably aspartic acid. The conformational separation provided by the amino acids of (B) 0 allows more efficient interactions between the specific immunogen presented and the appropriate Th cells and B cells and thus improves the immune responses to the Th epitope and the epitope that produces antibody and its cross reaction and immunologically functional homologs thereof. Th is a sequence of amino acids (natural or unnatural amino acids) comprising a Th epitope. A Th epitope can consist of a continuous or discontinuous epitope, hence not all Th amino acids are necessarily part of the epitope. Th epitopes, which include homologs and Th epitope segments, are capable of enhancing and stimulating an immune response to the CD4-CDR2 antigen peptides (eg, SEQ ID NOS: 4 and 5, and immunologically functional homologs of the same). Th epitopes that are immunodominant and promiscuous are highly and widely reactive in animal and human populations with widely divergent MHC types (Partidos et al., 1991; US 5,759,551). The Th domain of the subject peptides has from about 10 to about 50 amino acids and preferably from about 10 to about 30 amino acids. When multiple Th epitopes are present (ie, > 2), then each Th epitope is independently the same or different. The segments are contiguous portions of a Th epitope that are sufficient to enhance or stimulate an immune response to the CD4-CDR2 antigen peptides (for example SEQ ID NOS: 4 and 5), and / or to immunologically functional analogues of the same.
Th epitopes of the present invention include those derived from foreign pathogens and are provided as examples, but are not limited to, epitopes of helper T cell, hepatitis B surface and core antigen (HBS Th and HBC Th), cell epitopes T pertussis toxin assistant (PT Th), tetanus toxin assistant T cell epitopes (TT Th), T-cell epitopes assistant to measles virus F protein (MVF Th), T cell epitopes membrane protein assistant Main external of Chlamydia trachomatis (CT Th), diphtheria toxin assistant T cell epitopes (DT Th), attendant T-cell epitopes of Plasmodium falciparum circumsporozoite (PF Th), helper T-cell epitopes of Schistosoma triose phosphate isomerase mansoni (SM Th), and T-cell epitopes attendant Trat of Escherichia coli (Trat Th). Th epitopes derived from pathogens listed as SEC. FROM IDENT. US Nos. 2-9 and 42-52 in US Patent 5,759,551; as a helper T cell of Chlamydia P11 in Stagg et al., Immunology, 1993; 79; 1-9; and as a HBc peptide 50-69 in Ferrari et al., J Clin Invest, 1991; 88: 214-222; they are incorporated herein by reference and are listed herein along with others as SEC. FROM IDENT. NOS: 8, 13, 38-58 (Table 8). The epitopes additionally include artificialized Th for example, SEC. FROM IDENT. US: 6, 12, 36, 59 (Table 9), and immunologically functional homologs. Functional Th homologs include immunomodulatory homologs, cross-reaction homologs and segments of any of these Th epitopes. Functional Th homologs further include conservative substitutions, additions, deletions and insertions from one to about 10 amino acid residues and the epitope. Th which does not substantially modify the Th stimulating function of the Th epitope. The peptide conjugates of the invention also include CD4-CDR2 antigen peptides (eg, SEQ ID NO: 4 or 5) coupled to a carrier protein (e.g. , lamellated hemocyanin). Preferred peptide immunogens of this invention are peptides containing the CD4-CDR2 antigen peptides (eg, SEQ ID NO: 4 or 5, or immunologically functional homologs thereof) and Th epitopes, and optionally a general immunostimulator, for example, Inv (SEQ ID NO: 7). In a more preferred embodiment, the Th epitope is a HBS Th, HBC Th, MVF Th, PT Th, TT Th, CT Th or HIV Th derived from foreign pathogens or an idealized artificial Th, or a functional immunogenic homologue thereof. Optionally, A is a general immunostimulatory peptide, for example, Inv (SEQ ID NO: 7), preferably linked via a Gly-Gly or eNLys spacer. The structure of the modified site is based on a peptide sequence taken from the CDR2-like domain of human CD4 (amino acids 27-66 of SEQ ID NO: 1), or the homologous sequence of other species. This CD4-CDR2 target site is subjected to the following modifications: (1) the addition or insertion of a cysteine residue close to the N-terminus, (2) the addition or insertion of a cysteine residue near the C-terminus, preferably in, or near position 66 or a homologous position, and (3) the formation of a disulfide bond between the retained cysteines to produce a cyclic structure. Peptide structures may also comprise from 1 to 5 additional amino acids taken from any term in segment 27-66 or 39-66 of CD4, provided that the single undissolved cyclic disulfide structure (eg, SEC. IDENT. NOS: 10 and 11). Preferably, any cysteines intervening in the native sequence not intended to be employed for cyclization will be replaced conservatively by, for example, serine. For example, the target sites of human CD4-CDR2 (SEQ ID NOS: 2 and 3) are cyclized by adding cysteines in or near both N- and C-terminals (e.g.
SEC. FROM IDENT. NOS: 4 and 5) or through a cysteine added at the N-terminus and a cysteine substitution near the C terminus (for example, substituting Cys for Phe at position 67, to obtain SEQ ID NOS: 10 and 11). The modified CD4-CDR2 antigen peptides, cyclized and overlapped with the following sequences Cys His Trp Lys Asn Trp Asn Gln Me Lys and Leu Gly Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp Arg Ala Asp Ser Arg Arg Ser Leu Trp Asp Gln Gly Asn Cys (SEQ ID NO: 4) Cys Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp Asp Gln Gly Asn Cys (SEC. DE IDENT NO: 5) Cys His Trp Lys Asn Trp Asn Gln Me Lys Lie Leu Gly Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp Asp Gln Gly Asn Cys Pro Leu lie Me (NAME IDENTIFICATION SECTION) Cys Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp Asp Gln Gly Asn Cys Pro Leu lie Me (SEQ ID NO: 11) are provided as examples . In these examples, the modified positions are indicated by the bold type.
The antibody that is evoked by the peptide immunogens comprising the CD4-CDR2 antigen peptides of the invention is cross-reactive with the host cell receptor / co-receptor complex comprising CD4, and neutralizes primary HIV isolates . The corresponding target antigenic sites for the CD4 surface of other species can in the same way be derived from the homologous segments of the relevant species. The cross-reactive and immunologically functional homologs of the CD4-CDR2 antigen peptides (SEQ ID NOS: 4, 5, 10 and 11) may additionally comprise conservative substitutions, additions, deletions or insertions from one to about four amino acid residues. , with the proviso that the peptide homologs are capable of producing cross-reactive immune responses with the CD4-CDR2 peptides (SEQ ID NOS: 2, 3, 4 and 5) and that they have neutralizing activity against the isolates primary HIV Conservative substitutions, additions and insertions are known to those skilled in the art, and can be easily achieved with natural or non-natural amino acids as defined herein. Preferred peptide immunogens of this invention are peptides containing (1) the cyclic modified CD4-CDR2 sites referred to herein as CD4-CDR2 antigen peptides (eg, SEQ ID NOS: 4, 5, 10 and 11) or immunological homologs thereof and (2) Th epitope peptides. The most preferred peptide immunogens are those tandem constructions containing the cyclized CD4-CDR2 antigen peptides (SEQ ID NOS: 4, 5, 10 and 11) or cross-reactive and immunologically functional homologs thereof; a spacer (for example, Gly-Gly or eNLys); a Th epitope selected from the group consisting of a HBS Th, HBC Th, MVF Th, PT Th, TT Th, SMTh, HIVTh (eg, SEQ ID NO: 8, 38-50, 55), Th artificial (e.g., SEQ ID NOS: 6, 12, 36, 59), or a homolog thereof; and optionally, an Inv domain (SEQ ID NO: 7) or analogous thereof. The peptide immunogens of this invention can be manufactured by chemical synthesis methods which are well known to commonly skilled artisans. See, for example, Fields et al., Chapter 3 in Synthetic Peptides: A User's Guide, ed. Grant, W.H. Freeman & Co., New York, NY, 1992, p.77. Hence, the peptides can be synthesized using the automated Merrifield techniques of solid phase synthesis with the α-NH2 protected by either t-Boc or Fmoc chemistry, and using commercially available protected side chain amino acids. Examples of suitable instruments for peptide synthesis are the Applied Biosystems Peptide Synthesizer Model 430A or 431.
After complete assembly of the desired peptide immunogen, the resin is treated according to standard procedures to unfold the peptides from the resin and unblock the functional groups on the amino acid side chains. The free peptide is purified by HPLC and characterized biochemically, for example, by amino acid analysis, by mass spectrometry, and / or by sequencing. The methods of purification and characterization for the peptides are well known to those skilled in the art. Other chemical means for generating the peptide constructs of the invention containing CD4 and the Th sites include the linking of haloacetylated and cysteinized peptide through the formation of a "thioether" bond. For example, a cysteine can be added to the C terminus of a peptide containing Th and the thiol group of the cysteine can be used to form a covalent bond for an electrophilic group (for example, an N- (chloroacetyl) or a maleimide derived with an a- or e-NH2 group of a lysine residue) linked to the N terminus of a CD4-CDR2 antigen peptide. Thus, a conjugated composition is peptide comprising Th- (B) 0- (CD4-CDR2 peptide antigen) or its inverse, (peptide antigen CD4-CDR2) - (B) 0-Th, with or without general immunostimulatory site , can be obtained, where one of the binders "B" is Gly-Gly, (eN) Lys, -NHCH (X) CH2SCH2CO-, -NHCH (X) CH2SCH2CO (eN) Lys-, -NHCH (X) CH2S- succinimidyl (eN) Lys-, or -NHCH (X) CH2S- (succinimidyl) -.
The immunogenic subject can also be polymerized. The polymerization can be achieved by the reaction of the immunogen with a cross-linking reagent, for example by reaction between the glutaraldehyde and -NH2 groups of the lysine residues, using routine methodology. By other methods, the synthetic peptide immunogens can be polymerized or co-polymerized by utilizing an additional cysteine added to the N-terminus of the immunogen. The thiol group of the N-terminal cysteine can be used for the formation of a "thioether" bond with amino acid-modified haloacetyl or a maleimide derived with an a- or e-NH2 group of a lysine residue that binds to the N -terminal of a molecule of branched poly-lysyl nuclei (for example, K2K, K4K2K or K8K K2K). The immunogenic subject can also be polymerized as a branched structure through the synthesis of the desired peptide construct directly onto a branched poly-lysyl core resin (Wang, et al., Science, 1991; 254: 285-288). Alternatively, the longer synthetic peptide immunogens can be synthesized by well-known recombinant DNA techniques. Many standard DNA technology manuals provide detailed protocols for producing the peptides of the invention. To construct a gene encoding a peptide of this invention, the amino acid sequence is translated inversely within a nucleic acid sequence, preferably using the codon treatment used for the organism in which the gene will be expressed. Next, a synthetic gene is made, typically by synthesizing overlapping oligonucleotides which encode the peptide and any necessary regulatory elements. The synthetic gene is inserted into a suitable cloning vector and the recombinants are obtained and characterized. The peptide is then expressed under suitable conditions appropriate for the selected expression system and the host. The peptide is purified and characterized by standard methods. The nucleic acids described above may be useful in themselves as components of the so-called "DNA vaccines". In this embodiment of the invention, the expression of the immunogenic peptides of the invention is induced in the patient's own cells, by introducing into those nucleic acid cells encoding the peptides, preferably using codons and promoters that optimize expression in human cells Methods for making and using DNA vaccines are described in U.S. Patent Nos. 5,580,859, 5,589,466, and 5,703,055; see also WO 97/02840 and W. McDonnell and F. Askari, New Engl. J. Med., 1996, 334: 2-45, all of which are incorporated herein by reference. Such methods for making and using the peptides and peptide conjugates of this invention are contemplated to be within the scope of this invention.
The efficiency of the peptide composition of the present invention can be established by injecting animals, eg, guinea pigs, followed by observation of the humoral immune response to the CD4-CDR2 antigen peptides by the ability of the immune serum to neutralize primary HIV isolates as detailed in the Examples. Another aspect of this invention provides a vaccine composition comprising an immunologically effective amount of one or more of the peptide immunogens of this invention in a pharmaceutically acceptable delivery system. Such immunogenic compositions are used for the prevention and treatment of the immunodeficiency virus infection, as well as for the treatment of undesirable immune responses mediated by CD4 expressing T cells, such as transplant rejection, and autoimmune disorders such as rheumatoid arthritis, lupus. systemic erythematosus and psoriasis. Accordingly, the peptide composition of the invention can be formulated as an immunogenic composition using adjuvants, emulsifiers, pharmaceutically acceptable carriers or other ingredients routinely provided in vaccine compositions. Adjuvants or emulsifiers that can be used in this invention include alum, incomplete Freund's adjuvant, liposine, saponin, squalene, L121, emulsifier, monophosphoryl lipid A (MPL), dimethyldioctadecylammonium bromide (DDA), QS21, ISA206 and ISA 720 , as well as other known effective adjuvants and emulsifiers. Such formulations are readily determined by one of ordinary skill in the art and also include formulations for immediate release and / or sustained release, and for the induction of systemic immunity and / or induction of localized mucosal immunity, which can be achieved, for example, by immunogenic capture, or by co-administration with microparticles. The present vaccines can be administered or any convenient route that includes subcutaneous, oral, intramuscular route, or some parenteral or enteral route. Similarly, immunogens can be administered as a single dose or multiple doses. Immunization schedules are easily determined by the common expert artisan. The immunogenic composition of the present invention contains an effective amount of one or more of the peptide immunogens of the present invention and a pharmaceutically acceptable carrier. Such a composition in a suitable unit dosage form generally contains about 0.5 μg to about 1 mg of the immunogen per kg of body weight. When supplied in multiple doses, it can be conveniently divided into an appropriate amount per unit dosage form. For example, the initial dose, for example, 0.2-2.5 mg; preferably 1 mg, of immunogen represented as a peptide composition of the present invention, will be administered by injection, preferably intramuscularly, followed by repeated doses (booster). The dosage will depend on the age, weight and general health of the patient as is well known in therapeutic vaccine techniques. Vaccines containing mixtures of the peptide immunogens of the subject with two or more Th epitopes can enhance immunoefficiency in a larger population and thus provide an improved immune response to the CD4-CDR2 antigen peptide (e.g., SEQ ID NOS. : 4 and 5). The immune response to the synthetic CD4-CDR2 immunogens of the invention can be improved by delivery through capture in or on biodegradable microparticles of the type described by O'Hagan et al. (Vaccine, 1991; 9: 768). Immunogens can encapsulate with and without an adjuvant, and such microparticles can carry an immunostimulatory adjuvant. The microparticles can also be co-administered with peptide immunogens to enhance immune responses that include localized mucosal immunity which can be applied especially to a mucosal-mediated virus such as HIV, and to provide controlled release over time for sustained or periodic responses, for oral administration, and for local administration. (O'Hagan et al., 1991; and, Eldridge et al., 1991; 28: 287).
For this invention to be better understood, the following examples are set forth. These examples are for illustrative purposes only, and should not be construed as limiting the scope of the invention in any way.
EXAMPLES The target antigenic site peptides of these Examples were synthesized by the solid phase method underlined in Example 1. Each peptide can be represented by the formula (A) n- (Th) m- (B) 0- (CD4-CDR2 peptide antigen) or (A) n- (CD4-CDR2 peptide antigen) - (B) 0- (Th) m, but other formulas as described above are also encompassed within the invention. The target antigenic site CD4 is a cyclized peptide, exemplified by SEC. FROM IDENT. NOS: 4, 5, 10 and 11, but immunologically functional homologs comprising a cyclic peptide in the range of about 25 to about 50 amino acids, having an contiguous amino acid sequence derived from SEC. FROM IDENT. NOS: 2 or 3 and up to a sequence of five additional amino acids linked to the N-or C-terminal of the cyclic structure, are intended to be within the scope of the invention. Each peptide using for these examples has Gly-Gly or eNLys as the spacer (B) 0 between the Th and the immunogenic elements of the modified CD4-CDR2 target site and some incorporate an optional element (A) 3 comprising Inv-Gly-Gly wherein Inv (SEQ ID NO: 7) is coupled to the antigenic peptide (e.g., SEQ ID NOS: 32-35), although peptides of the invention may also have other spacers (e.g. IDENTIFICATION NO: 9, eNLys) or non-spacers. The Th epitopes, as exemplified in Table 8, include promiscuous helper sites derived from foreign pathogens such as measles virus F protein and hepatitis B virus core and surface, and other Th epitopes as shown in Table 8 ( SEQ ID NOS: 8, 13 and 43-58) and artificial Th is as shown in Table 9 (for example, SEQ ID NOS: 6, 12, 36 and 59). The peptides of this example also include an optional general immunostimulatory site (eg, SEQ ID NO: 7). In addition, the invention is not limited to Inv as the additional immunostimulatory element.
EXAMPLE 1 IDENTIFICATION OF POTENTIAL EFFECTORS SITES ON THE CD4 / CO-RECEPTOR SURFACE COMPLEX A. Peptide Design The sites within all four domains of human CD4 together with the co-receptor sites representing the four external domains of human CD4 receptors. chemokines, which include CC-CKR1, CC-CKR2b, CC-CKR3, CCKR5 and LESTR were selected for peptide mimicry. As the "epitope" recognized by MAb B4 (WO 97/46697) is conformational in nature, none of the linear peptides derived from the above receptor / co-receptors reacted strongly with MAb B4, although the reactivity of MAb B4 with rsCD4 was significantly improved in the presence of certain peptides derived from the co-receptor domains of chemokine as shown in WO 97/46697. Despite the lack of strong reactivity of MAb B4 with any single CD4- or peptides derived from chemokine co-receptor, weak MAb B4 reactivities were detected for peptides derived from various CD4 regions (AA1-A20, AA81-92, AA60- AA109, AA118-AA165, AA235-251, AA297-AA351, or AA361-AA375). This prompted a different response that aims to design synthetic peptides that would produce high-affinity antibodies reactive with one or several conformational-like sites recognized by MAb B4, for inhibition of HIV infection of the target cells. The sequences of such potential sites disseminated through the four domains of CD4 and the external domains of various chemokine co-receptors were therefore designed and synthesized as target peptides and rendered immunogenic by construction peptides wherein the promiscuous derivatives of HBsAg (SEQ ID NO: 8) and Inv (SEQ ID NO: 7) were linked to the target sites, as shown in Tables 1 and 2. The specific CD4 sites within these domains were selected for cyclization based on the predictions of Brookhaven's three-dimensional model for human CD4 (http: www.pdb.bnl.gov/pdb.bin). / pdbids) of undulations exposed to surfaces. The specified cyclic constraints were installed within these peptides to maximize cross-reactions between the target antigenic sites and the native CD4 molecule. Accordingly, several of the synthetic constructs of Tables 1 and 2 were synthesized with introduced cysteines not found in the native sequence, to produce disulfide bond ripples to mimic the wave structures predicted by the Brookhaven model. In some cases, the naturally occurring cysteines were replaced with serines to prevent the formation of conformations not favored by the model. For peptides derived from a chemokine co-receptor, the cross-linking between the peptides of external domains 2 and 3, shown at the far right of Table 2, was made by means of naturally occurring cysteine residues in the respective domains, imitation of its native structure. The sites marked by * in the description column of Table 1 have been designed with a specified cyclization. Other peptide sites are linear. Peptides labeled "a" in the form columns of Tables 1 and 2 represent the CD4 or CCKR target antigen site alone. These were used as the substrate antigens for the ELISA-based peptide. Peptides labeled "b" were synthesized as target antigenic sites in tandem with the Th HBs site (SEQ ID NO: 8) as shown. Peptides marked by "c" are variants of the "b" constructs synthesized in tandem with the immunostimulatory peptide of the Inv domain (SEQ ID NO: 7) as shown in Tables 1 and 2. Peptides designed as " d "are variants of the" b "constructs synthesized in tandem with a second peptide Th CT P11 Th (SEQ ID NO: 13) linked to the N-terminus via a Gly-Gly linker. The peptides marked by "e" were synthesized as the inverse of "b" with the Th sites located at the C terminal and the target antigenic site at the N terminus of the construct. A peptide marked by "g" represents a branched tetrameric peptide with synthesis conducted directly on a resin nucleus. Peptides marked by "x" represent peptides comprising a two-chain structure linked by an inter-disulfide bond via the naturally occurring cysteine residues present on the respective chains. Other sites Th used in the experiments shown in Tables 1 and 2, but not shown here, employ the artificial Th sites, "1,4,9 PALINDROMIC" (SEQ ID NO: 6) and "Syn Th (1). , 2,4) "(SEQ ID NO: 12). The peptides with the Inv site located at the C terminus, and the CD4-CDR2 antigen at the N terminus (peptide antigen CD4-CDR2 GG-Th-GG-lnv) were also prepared, but are not shown.
Immunogenic peptides "b", "c", "d", "e", "x", and "others" Th used for the studies in Tables 1 and 2 were also synthesized with Gly-Gly spacers for separation of the target site antigenic site Th, and the Th removal of Inv or a second Th immunostimulatory site. The resulting peptide immunogens were selected as candidate antigenic sites for their ability to induce immunized host antibodies with the following properties: 1. Linkage to the target antigenic site in an ELISA test; 2. Link to rsCD4 in an ELISA test, in cases of antigenic peptides derived from CD4; 3. Link in an immunofluorescent test to T cells expressing the cell surface receptor / co-receptor complex comprising CD4; and 4. Primary isolates of HIV resistant to neutralizing neutralization in an in vitro microplate test. B. Selection of Antigenic Peptides Candidate Objectives: 1. Synthesis of Antigenic Peptides Objectives derived from Chemokine receptor and CD4. The peptides listed in Tables 1 and 2 in their corresponding form "a", "b", "c", "d", "e", or "x" were synthesized individually by Merrifield solid phase synthesis technique in automated peptide synthesizers from Applied Biosystems (Models 430, 431 and 433A) using Fmoc chemistry. The preparation of peptide immunogens comprises a library of structured synthetic antigen (SSAL) for artificial T cell epitopes "(1, 4, 9 PALINDROMIC) Th" (SEQ ID NO: 6) as achieved by providing a mixture of alternative amino acids for coupling at a given variable position, in the appropriate ratio as specified in the SEC design. FROM IDENT. NO 6. SSAL peptides that have library designs for B cell target antigen sites or other Th SSAL sites can be synthesized in a similar manner. After complete assembly of the desired peptide, the resin was treated according to the standard procedure using trifluoroacetic acid to unfold the peptide from the resin and unblock the protecting groups on the secondary amino acid chains. For the cyclic peptides, the split peptide was allowed to remain in 15% DMSO in water for 48 hours to facilitate the formation of the intrachain disulfide bond between cysteines. The cleaved, extracted and washed peptides were purified by HPLC and characterized by mass spectrometry and reverse phase HPLC. 1. The generation of Specific Immune Serum for the Target Antigenic Site derived from the Chemokine Receptor and CD4 for Functional Efficiency and Evaluation.
The immunogenic efficiency of the peptide compositions was evaluated as specified by the experimental immunization protocol delineated subsequently followed by serological antibody response tests. Standard Experimental Design: Immunogens: (1) Individual peptide immunogen; or (2) a mixture comprising an equimolar ratio of peptide immunogens as specified in each protocol. Dosage: 100 μg in 0.5 ml per immunization unless otherwise specified. Route: intramuscular unless otherwise specified. Adjuvants: (1) Complete Freund's Adjuvant (CFA) / Incomplete Adjuvant (IFA): (2) 0.4% Alum (aluminum hydroxide); or (3) other adjuvants as specified. One adjuvant per immunogen per group. Dosage Itinerary: 0, 2 and 4 weeks; or 0, 3 and 6 weeks; or as otherwise specified. The CFA / IFA groups received CFA week 0, and IFA in subsequent weeks. Alumbres or other specified adjuvant groups received the same formulations for all doses. Bleeding Schedule: Weeks 0, 3, 6 and 8, or as otherwise specified Species: Duncan Hartley Guinea Pigs Group Size: 3 guinea pigs / group Test: Specific ELISA for each antiseptic activity of immune serum. The solid phase substrates were those corresponding to the "a" form of the target antigenic peptide (e.g., target antigenic peptide CD4, peptide derived from the chemokine receptor, etc.). The blood was collected and processed in serum, and stored before titration by ELISA with the target antigenic peptides. 2. Serum and Antibodies The following serological reagents, derived from guinea pig immune serum, or murine or humanized monoclonal antibody, were used for evaluations in various serological tests. All sera from guinea pigs directed against rsCD4, and objective antigenic sites derived from chemokine and CD4 co-receptor were obtained as described above at various time points after immunization. Other serological reagents were obtained through previous studies or from external sources as described. These were incorporated occasionally for comparison purposes. For example, gp anti-gp120 V3 MN (anti-V3 MN) is pooled serum of guinea pigs that has been hyperimmunized with a synthetic peptide antigen corresponding to the hypervariable V3 domain of gp120 of HIV-1 MN (Wang et al., Science, 1991, 254: 285-288). The anti-gp120 V3 GP library serum is pooled antiserum from three guinea pigs hyperimmunized with a complex mixture of peptides representing an SSAL of approximately 1013 possible HIV-1 V3 sequences (anti-V3 SSAL). The V3 MN and V3 SSAL immunogens used for guinea pig immunizations were multiradified V3 synthetic peptide immunogens that were used to generate polyclonal antibodies with neutralizing activity for various HIV-1 laboratory strains as described in Walfield et al. (Chapter 18 ¡n AIDS Research Reviews, ed. Koff et al., Marcel Dekker: New York, 1993, pp.345-360).
Another anti-gp120 antibody was a recombinant human monoclonal antibody designated IgG 1 b12 with specificity for the gp120 binding site for CD4 (anti-gp120 CD4-BS) (Burton et al., Science, 1994, 266: 1024-1027). IgG1 b12 was generated as a Fab fragment from a library that presents phage antibody prepared from bone marrow from a long-term asymptomatic HIV-1 seropositive donor and converted to a complete human antibody by cloning into a DNA expression vector recombinant IgG1. It is considered as the "gold standard" of antibodies for neutralization of several primary isolates of HIV (Burton et al., Supra). 3. ELISA Anti-peptide. The anti-peptide antibody activities were determined by ELISA (enzyme-linked immunosorbent assays) using 96 well flat bottom microtiter plates which were coated with the corresponding target antigenic peptide in "a" form as an immunosorbent. Aliquots (100 μl) of a target antigenic peptide solution in a concentration of 5 μg / ml were incubated for 1 hour at 37 ° C. The plates were blocked by another incubation at 37 ° C for 1 hour with a 3% solution of gelatin / PBS. The blocking plates were then dried and used for the test. Aliquots (100 μL) of the test immune serum, starting with a 1: 100 dilution in a sample dilution regulator and serial dilutions ten times thereafter, were added to the peptide coated plates. The plates were incubated for 1 hour at 37 ° C. The plates were washed six times with 0.05% PBS / TWEEN® regulator. 100 μL of horseradish peroxidase labeled goat-anti-specific antibody species was added at appropriate dilutions in conjugate dilution buffer (Phosphate buffer containing 0.5M NaCl, and normal goat serum). Plates were incubated for 1 hour at 37 ° C before being washed as above. Aliquots (100 μL of o-phenylenediamine substrate solution were then added.) The color was allowed to develop for 5-15 minutes before the color enzymatic reaction was stopped by the addition of 50 μL of 2N H2SO4. from each well was read on a plate reader.The ELISA titers, shown as Log10 of reciprocal dilution, were calculated based on the linear regression analysis of the absorbances, with cut of A492 set at 0.5. rigorous since the values for the normal guinea pig control samples diluted runs with each test was less than 0.15 4. Determination of antibody reactivities with rsCD4, and with cells expressing CD4 a.Determination of anti-CD4 reactivity by rsCD4 ELISA Purified recombinant soluble CD4 (rsCD4) was obtained from a commercial source (American Bio-Technologies, Inc.
Cambridge, MA) and NIH (USA) AIDS Research and Reference Reagent Program. RsCD4 ELISA was conducted by coating 96-well microtitre plates by overnight incubation at 4 ° C with rsCD4 at 0.25 μg / ml using 100 μL per well in 10 mM NaHCO3 buffer, pH 9.5. Wells coated with rsCD4 were incubated with 250 μL of 3 wt% gelatin in PBS at 37 ° C for 1 hour to block non-specific protein binding sites, washed three times with PBS containing 0.05 volume% TWEEN 20 and then dried. Immune serum or monoclonal antibodies were serially diluted with PBS containing 20% by volume of normal goat serum, 1% by weight of gelatin and 0.05% by volume of TWEEN 20 at 1:20 volume by volume dilutions unless Indicate otherwise. 100 μL of the diluted sample was added to each of the wells and allowed to react for 1 hour at 37 ° C. The wells were then washed six times with 0.05% by volume of TWEEN 20 in PBS to remove labeled unbound antibodies. 100 μL horseradish peroxidase labeled goat anti-mouse IgG or goat anti-guinea pig IgG at a dilution of 1: 1000 in 1% by volume of normal goat serum, 0.05% by volume of TWEEN 20 in PBS was added to each well and incubated at 37 ° C for 15 minutes. The wells were washed six times with 0.05% by volume of TWEEN 20 in PBS to remove unbound labeled antibody conjugate and reacted with 100 μL of the substrate mixture containing 0.04% by weight of orthophenylenediamine (OPD) and 0.12% in volume of hydrogen peroxide in sodium citrate buffer pH 5.0, for 15 minutes. The reactions were stopped by the addition of 100 μL of 1.0 M H2SO4 and the absorbance at 492 nm (A492) was measured. The reciprocal Log10 antibody titer was calculated for the endpoint reactivity of each test sample, as interpolated by linear regression, as described for the anti-peptide ELISA. to. Determination of the reactivity of CD4 expressing cells by indirect immunofluorescent staining 0.5 x 10 6 cells expressing CD4 (e.g. cell line cells HPB-ALL, MT2 or SUP-T1) per well were washed twice in PBS containing 1% BSA before incubation with the designated immune serum or monoclonal antibodies, at an optimal concentration as determined for each experiment, for 45 minutes at room temperature. After incubation of the cells with the first staining antibody, the cells were washed for an additional two times in the same washing buffer and incubated with a secondary residue reagent of fluorescein isothiocyanate (FITC) conjugated goat anti mouse IgG or (F? TC) -conjugated goat anti-species specific IgG at appropriate dilutions (Cappel, Malvern PA) for an additional 45 minutes at room temperature. The stained cells were washed again in the same washing buffer and the cells were processed for fluorescence analysis by cytofluorography and / or immunofluorescence microscopy for the determination of the percentage of stained cells, and the intensity of the staining. b. Indirect test of immunofluorescence inhibition. For competitive link inhibition tests "Biotinylated monoclonal B4 antibody T cells" using the indirect immunofluorescence staining technique, the cells were first incubated with the interfering reagents or the appropriately diluted immune sera and washed twice in the same wash buffer before the addition of the biotinylated monoclonal antibody B4. The staining of T cells expressing CD4 was completed by subsequent incubation with Appropriately diluted FITC-avidin followed by three additional washes before analysis by high-resolution fluorescence microscopy or cytofluorography. 5. Determination of virus neutralization by antibodies. to. Cells The human T-cell MT-2 line (ATCC 237) was maintained in Dulbecco's modified Eagle medium supplemented with 15% fetal bovine serum as previously described (Hanson et al., J Clin Microbiol, 1990, 28: 2030- 2034). Peripheral blood mononuclear cells (PBMCs) were isolated from HIV-1 seronegative donors from fresh yellow leukocyte cover units by Ficoll-Hypaque separation gradient (Organon Teknika S.A., Durham, NC). The resulting PBMCs were stimulated with 0.5% PHA-P (Difco Laboratories, Detroit, Ml). After 3 to 4 days, the medium containing PHA-P was removed and the cells were maintained in RPMI with 15% fetal bovine serum, 900 μg / ml glutamine, antibiotics, and 5% interleukin-2 (Cellular Products , Inc., Buffalo, NY). b. Virus HIV-1 MN is an ATC strain available as and maintained as a persistently infected culture of H9 cells from the National Institute of Healt, Bethesda MD (NIH AIDS Research and Reference Reagent Program Catalog No. 402) from which free, concentrated standards were prepared of cells. Primary piles of HIV-1 were prepared from PBMC patients by co-cultivation PBMC. Standard cultures of primary isolates were prepared by no more than 3-5 passages through PBMCs, and clarified by centrifugation (Sawer et al., J Virol, 1994, 68: 1342-1349). They were supplied by Cari Hanson of the California Department of Healt Services, Berkeley CA. c. MT-2 Microplate Neutralization Test The determination of the antibody titre that neutralizes HIV employs the preincubation of serum or antibody diluted serially with a fixed amount of HIV followed by infection of HIV-sensitive MT-2 cells and the formation of a monocellular layer that presents HIV-induced microplates. The results were counted by quantification of the microplates. The test is suitable only for SI isolates, whether TCLA isolates or primary isolates, because the microplates represent giant syncytia formed by MT-2 cells that fuse to the foci of HIV-infected cells. The test is appropriate for evaluating the inhibition of virus-to-cell and cell-to-cell transmission because the inhibition of syncytium formation results from the action of the antibody on HIV particles or HIV-infected cells, ie the test measures the inhibition of induced HIV fusion from virus to cell or induced fusion of HIV from cell to cell. The neutralization is then observed by the reduction of microplates as observed by the enumeration of the plates stained with propidium iodide 1 week later (See, Hanson et al., J Clin Microbiol, 1990, 28: 2030-2034). In this test, the virus and serum or antibody were diluted 50% in normal human defibrinated plasma pooled to negate any non-specific increases or inhibitory effects. C. Results: Peptides and objective antigenic sites derived from candidate chemokine or CD4 co-receptor used for immunogenicity and preliminary functional studies are described in Tables 1 and 2. The guinea pigs were immunized as described above with the "b" forms or "c" of the target antigenic site unless otherwise indicated in Tables 3 and 4, and the immune serum collected 6 or 8 weeks after the initial immunization was analyzed by antiseptide ELISA and rsCD4 ELISA as described in Procedures As shown in Tables 3 and 4, most peptide immunogens derived from chemokine and CD4 co-receptor were highly immunogenic as they evoked antypeptide antibodies with titers in the range of 2.5 a >; 5 Log10, except for peptides p1590b, p1699b, p1699c and p1700b. Antigenic sites derived from CD4 comprising long segments of the CD4 receptor (eg, p1612c, p1678b, p1678c, p1686b, p1697b, p1817b, p1889b and 1901b) together with some cyclized target sites were high cross reacted with rsCD4, as shown by their corresponding titles > 3.5 Log10 by the anti-rsCD4 ELISA (see Table 3, column A2). The cross-reactivity with rsCD4 for each of the peptide constructs was not predictable. In addition, such cross-reactivity of rsCD4 did not extend to the CD4 surface of the corresponding host cell because among those peptide constructs having high cross-reactivity of rsCD4, only p1697b and p1901b were found to be strongly reactive with T cells expressing CD4 by staining indirect immunofluorescence with cells of the cell line HPB-ALL or MT2 (Table 3, column B). In contrast, serum derived from CD4 target antigenic peptide sites with a cyclized structure (eg p1472b, p1472c) or from the CDR2 domain (eg, p1403b, p1471c) were highly reactive with CD4 expressing T cells despite their low cross-reactivity with rsCD4 (Table 3, column B). For the chemokine co-receptors, the sera derived from peptide constructs p1990, p1999, p2028, p2047, p2048, p2049, p2087 and p2089, the majority consequence of domains 1, 3 or 4 of the co-receptors, were found reactive with the "receptor / co-receptor surface complex" (Table 4, column B). The above results indicate that the cross-reactivity with rsCD4 or receptor surface complex / co-receptor is a complex and unpredictable phenomenon, influenced by conformational characteristics that can only be deduced by experimental observation. The immune serum (6 or 8 weeks after the initial immunization) obtained by the above peptide constructs was also selected for its neutralizing activity against a VL 135 isolate of HIV-1 from Ciado B by the microplate neutralization test MT- 2 as described above. Despite the presence of high titer cross-reactivity antibodies with rsCD4 or "CD4 surface complex / co-receptor" in some of the immune sera, none showed significant levels of such neutralizing antibodies (Tables 3 and 4, column C) . Immune sera that have bright immunofluorescence staining patterns with T cells expressing CD4 were further evaluated for their ability to inhibit or block binding by B4 MAb to T cells expressing CD4 to locate potential effector sites in proximity to discontinuous sites of the conformational epitope recognized by MAb B4. The results obtained from such experiments can lead to views for the effective design of new peptide immunogens. This evaluation was achieved by experiments involving the inhibition of immunofluorescence staining of the binding of "T cells to MAb B4-T". CD4 + target T cells (e.g., MT2 T cells) were reincubated with appropriately diluted immune sera (e.g., 1:10) followed by incubation of the cells with biotinylated avidin MAb B4 and FITC conjugate with detailed procedures described above. Among all the immune sera evaluated, only the sera generated through immunization with the peptide p1471c of the CD4-CDR2 domain was found to be inhibitory of the MAb B4 linkage (Table 5). None of these immunizations with peptides derived from chemokine co-receptor interfered with the binding of "T cells to MAb B4". This lack of inhibition of links from "T cells to MAb B4" may be related in part to less than optimal affinity presented by the antibodies towards the potential effector sites, and may not be exclusively due to the spatial distance of the sites represented by the target antigenic sites to that recognized by MAb B4.
All but one of the hyperimmune sera targeted against the receptor and co-receptor peptides failed to inhibit MAb B4 binding in T cells, and none exhibited neutralizing activity against a primary HIV isolate. Additional attempts were made to design new peptide constructs for the purpose of capturing the potential effector sites on the surface of the CD4 molecule, based on the position of p1471 in the CD4 sequence, the clue provided by the inhibition study. Binding of T cells to MAb B4-T ". More specifically, peptides comprising target antigenic sites surrounding the CD4-CDR2 domain spanning amino acid residues from 20 to 75 according to the numbering system of SEC. FROM IDENT. NO: 1 were revisited and additional peptide constructions covering this region were redesigned with particular emphasis on the preservation of the three-dimensional structure of this region by insertion of cysteine residues in both N- and C-terminal peptides derived from this CDR2 region with a ripple size in the range of 30 to 45 amino acids. The amino acid sequence for the representative peptide constructs derived from this region are shown in Table 6. The immune sera were collected at 6 or 8 weeks after the initial immunization and evaluated similarly to those described in the previous selection. Among the 41 target antigenic sites evaluated, p2057, p2189, p2190, and p2240 (SEQ ID NOS: 4, 11, 10, and 5), as "c" constructs (SEQ ID NOS: 32-35) , were found to produce neutralizing antibodies directed against primary HIV-1 isolates (Table 6). Isolate VL135 (Table 6) (Sawyer et al., J. Virol., 1994, 68: 1342-1349) is a primary isolate resistant to representative neutralization. It is not a primary isolate sensitive to atypical neutralization that can be used to provide apparent but misleading positive (D. Burton and J. Moore, Nature Medicine, 1998, 4: 495-48). Therefore, the viral neutralization observed herein, is not the inactivation of an easily neutralized virus but is evidence of protective immunity of a challenge by an isolated HIV field. There have been no previous observations in the field of AIDS research where chemically defined immunogens produced antibodies with this critical HIV neutralization function. Despite the lack of binding activity by MAb B4 to any of the peptide CD4-CDR2 domain sites (as shown in the co-pending patent application published as WO 97/46697), it is demonstrated here that a CD4 target site -CDR2 is proximal to the disseminated discontinuous epitope that constitutes the recognition site for the neutralizing monoclonal antibody B4. This recognition site appears to contain peptide sites from all four CD4 domains probably due to the curved nature of the "receptor surface / co-receptor complex comprising CD4", recognized by MAb B4. This "surface CD4" is different from the well-known extended 3D model of rsCD4. It is noteworthy to note that only certain target antigenic sites derived from the CD4-CDR2 region, for example, those spanning a more widespread area presented as cyclic peptides (e.g. p2057c, p2189c, p2190c and p2240c, SEQ ID NOS: 32-35), produced such neutralizing antibodies EXAMPLE 2 HIPERINMUNES SERIES GENERATED BY THE PEPTIDES p2057c AND p2240c DEMONSTRATED ANTIBODIES NEUTRALIZERS EXTENDINGLY REACTIVE AGAINST HIV PRIMARY MULTIPLE CLAPS As shown in Table 7, the hyperimmune sera derived from the indentations obtained 15 and 12 weeks after the initial immunization for peptide immunogens p2057c and p2240c (SEQ ID NOS. : 32 and 35) demonstrated significant neutralizing 90% antibody titers against primary HIV isolates from multiple cies in a pattern parallel to that demonstrated by MAb B4. Neutralizing antibody titres varied in an increasing order from 1:20 to 1: 185 for primary isolates of all D, A, B (DH12), E, a B (VL 135) and C a C for immune sera for p2057c (SEC DE IDENT NO: 32); from 1:20 to 1: 324 for primary isolates of all D, B (DH12), A, E a ciated B (VL 135) for immune sera for p2240c (SEQ ID NO: 35). For the purpose of determining equivalence, the neutralization activity of MAb B4 varied from 25.6 μg / ml to 1.54 μg / ml for primary isolates of HIV-1 from all D, A, E, C, B (DH12) B (primary isolate VL 135). In comparison, guinea-pig sera directed to the N-terminal gp120 V3 domain and monoclonal antibodies directed to the N-terminal gp120 V3 domain of HIV-1 MN (MAb 50.1) or a less variable conformational CD4 gp120 binding site (lgG1 b12 MAb) failed to neutralize any of these primary HIV isolates resistant to neutralization. Dilutions from 1:20 to 1: 300 of the anti-p2057c and anti-p2240c immune sera provided 90% neutralization of primary HIV isolates from all A to E, as shown in Table 7, would be approximately one concentration of MAb B4 in undiluted serum of approximately 300 μg / ml of a calculation of MAb B4 concentration and serum dilutions in equivalent neutralization activities (ie an average of the MAb B4 concentration (μg / ml) x factor of dilution of immune serum for neutralization activity against each of the corresponding isolates).
EXAMPLE 3 CORRELATION OF NEUTRALIZING ANTIBODY TITLES IN SERUM WITH PROTECTION EFFICIENCY IN MACACOS OF INDIA AGAINST INVENTION OF SIVmac251 In co-pending patent application WO 97/46697, the protective efficiency of MAb B4 and its correlation to in vitro neutralization antibody titers was evaluated by a MAb challenge test B4 against the experimental infection of the Indian macaques with SIV, an animal model commonly used for human AIDS. In that study, MAb B4 was introduced to macaques from India and the serum was collected in pre-treatment, pre-challenge, and 1 hour after challenge for evaluation. The serum level of MAb B4 antibody, determined by immunoassay of rsCD4 against a pre-calibrated MAb B4 curve, was found to be in the range of 30-45 μg / ml for all animals that received a dosage of 5 mg / kg of body weight. Three of four animals that received this amount MAb B4 were protected against SIVmac 251. during the observed one year period. The level (ie, 30-45 μg / ml) of B4 MAb serum present in challenged animals is much lower than the estimated level of antibody (CD4-CDR2 antigen peptide) (around 300 μg / ml) that would be present in immune sera generated in the hosts if they had been immunized with an immunogenic composition comprising p2057c or p2240c (SEQ ID NOS: 32 and 35). Therefore, the hosts immunized with a peptide composition of the present invention comprising SEC. FROM IDENT. NOS: 4, 5, 10 and 11 or homologs thereof can be predicted to have protective immunity of VI H infection by primary VI H isolates from multiple ciados.
EXAMPLE 4 IMMUNOGENIC PEPTIDE COMPOSITIONS COMPRISING AN EPITHOPE Th ARTIFICIAL PROMISCUO An artificially designed Th / CD4-CDR2 antigen peptide was synthesized (SEQ ID NO: 6) -Gly-Gly- (SEQ ID NO: 5) and given the designation SEC. FROM IDENT. NO: 60 The peptide of SEC. FROM IDENT. NO: 60 was formulated in ISA 206 / DDA. ISA 206 / DDA is an oil / water emulsion in which MONTANIDE ™ ISA 206 is dispersed at 30 mg / ml (MONTANIDE ™ ISA 206 is an oily metabolizable solution supplied by SEPPIC Inc. of Fairfield, NJ). The oil suspension was then emulsified at a volume ratio of 1: 1 within an aqueous peptide solution which has been adjusted for a peptide content to provide the desired dose of peptide composition in 0.5 ml of the final preparation. The immunogenicity of SEC. FROM IDENT. NO: 60 in the above formulations was established in guinea pigs that received 100 μg / dose, given at weeks 0, 3, and 6. The immunogenicity was determined by antipeptide ELISA as described in Example 1 using SEC. FROM IDENT. NO: 5 as the cyclized target antigenic site peptide for the solid phase substrate. Six of six guinea pigs seroconverted successfully in ELISA reactivity. Significantly, the SEC. FROM IDENT. NO: 60 was also found to be highly immunogenic and functionally active in a large animal. An immunogenic composition comprising SEC. FROM IDENT. NO: 60 was formulated in Incomplete Freund's Adjuvant (IFA), 300 μg / dose, and administered to a pig by intramuscular injection at weeks 0, 3, and 6. The seroconverted pig and week 8 serum was tested by neutralization activity against primary isolate HIV-1 VL135 by the Microplate Neutralization Test of MT-2 (Example 1). The pig serum sample provided 50% neutralization of the feeding virus at a dilution of 1: 249, and 90% neutralization at a dilution of 1:97. Therefore, immunization with a peptide composition of the invention communicated to a large host animal with an immune response that includes antibodies to host cell receptors comprising CD4 and HIV neutralizing activity.
EXAMPLE 5 PEPTIDE CONSTRUCTIONS REPRESENTATIVE OF THE INVENTION The immunogenic peptides of the invention shown in Table 10 are completely synthetic constructs that were synthesized by the solid phase method set forth in Example 1. Each peptide in the Table can be represented by the formula (A) n- (Th) m - (B) 0- (CD4-CDR2 antigen) -X, but it is understood that the peptides of the other formulas described above are encompassed within the peptides of this invention. The sequence of CD4-CDR2 antigen is SEC. FROM IDENT. NO: 4 or 5. The immunogenic peptides shown comprise the artificial Th sites (as shown in Table 9). Each peptide of this example has Gly-Gly spacers between the immunogenic elements, but the peptides of the invention may have other spacers such as eNLys or without spacers. Materials and methods Representative peptide constructs of the invention as listed in Table 10 (SEQ ID.
Nos: 60, 61 and 62) were synthesized, unfolded, cyclized and purified, as described in Example 1. Peptide constructs were formulated for immunization within small animals such as guinea pigs, or within larger animals such as pigs or mandrels for evaluation of their immunogenicities. The peptides are suspended in a volume of 0.5 mL containing representative emulsifiers or adjuvants such as ISA51, ISA720, DDA or monophosphoryl lipid A (MPL). The dose was 100 μg of peptide for guinea pigs or 300 μg of peptide for pigs or baboons and the animals were immunized intramuscularly. The animals received injections at weeks 0, 3 and 6 as specified in Table 1. The test bleeds of 5, 8 or other specific weeks after the initial immunization were evaluated for cross-reactivity to rsCD4 by rsCD4 ELISA as described in Example 1, and were further tested for their ability to neutralize HIV-1 primary isolates also as described in Example 1. Results The representative peptide constructs were of relevant immunogenicity, since all the peptides tested produced strong cross-reactivity directed to the site to the corresponding rsCD4, as shown by the Log10 titers in the anti-human rsCD4 ELISA of more than 3.5 (Table 11). Neutralization of primary HIV-1 isolates (e.g. VL135) was also observed for immune sera obtained from guinea pigs, pigs, and baboons. This functional cross-reactivity for mandrel sera is worthy of attention so that the neutralization of primary isolates of human HIV by the mandrel serum is almost a human system. Thus, the efficiency of a peptide construct of the invention, as an agent for the prevention and / or immunotherapy of HIV infection by active immunization, is strongly indicated by the primate model. t The shape of peptide constructs is designated as a, b, c, e, g, and x, where: a represents an Antigenic Site Objective b represents an Antigenic Site Objective HBsTh-GG- c represents an Antigenic Site Target Inv-GG- HBsTh-GG- e represents an Objective Antigenic Site -GG-HBsTh g represents a branched tetramer of an Objective Antigenic Site on the core K2KAA x represents the crosslinking of peptide chains through an interchain disulfide bond. 10 * The peptide is cyclized through cysteines at or near N and C terms of the Target Antigenic Site Table 2 t The forms of the peptide constructs are en istan as a, b, d, e, and x, where: a represents an Antigenic Site Objective b represents an Antigenic Site Objective HBsTh-GG- d represents an Antigenic Site Target CT P11 Th- GG-HBsTh-GG- e represents an Antigenic Site Objective -GG-HBsTh x represents the cross-linking of two peptide chains through a disulfide linkage chain # peptides from the external domain of the chemokine receptor, numbering system of the amino acid sequences deducted from the nucleic acid sequences in: LESTR (Loetscher ef al, J. Biol. Chem. 1994, 269: 232) CC-CKR1, CC-CKR2b, CC-CKR3, CC-CKR5 (M. Samson et al, Biochemistry 1996, 35, 3362) Legend, Table 3: #: Conjugate p1460 - BSA A1: Reciprocal titre Log10 ELISA anti-target site A2: Title Log10 Reciprocal ELISA anti-rsCD4 B: IFA (Indirect Immunofluorescence staining test) C: Dilution of sera giving 50% of inhibition in the MT-2 neutralization test in Ciado B HIV-1 VL 135 g: Trimeric tetrameric peptide tr: trace 10 NT: not determined Table 4 Immunogenicity and Functional Mapping by ELISA Tests and Neutralization of Peptides Derived from the Chemokine Co-receptor A: Title Log10 ELISA anti-target site B IFA (Indirect Immunofluorescence Staining Test) C Dilution of serum giving 50% inhibition in the MT-2 Neutralization test in Ciado B HIV-1 VL 135 tr footprint Table 5 MAB B4 LINK INHIBITION TO MT2 T CELLS FOR IMMUNE SUMS GENERATED BY PEPTIDES DERIVED FROM CORRECEPTOR OF CHEMOCCAN OR CD4 t IFA # IFA inhibitory * see legend of Table 2 for the description of peptide codes 10 ± medium only A1: Title Log10 ELISA reciprocal antigenic site target B: IFA A2: Title Log 10 ELISA reciprocal anti-rsCD4 C: Dilution of serum that gives 50% inhibition in the Neutralization test of MT-2 in Ciado B HIV-1 VL 135 *: cyclized peptides Table 7 Neutralization of Primary Isolates of HIV-1 from Ciados A, B, C, D and E by Monoclonal Antibodies and Immune Serums (MT-2 Microplate Neutralization Test) #: HIV-1 primary isolates of HIV-1 provided by the WHO Global Program on AIDS.
Table 8 SEQUENCE OF AMINO ACIDS OF EPITOPES TH DERIVED FROM PATHOGENS STRANGE Ta lar Amino Acid Sequence of Artificial Th Epitopes Table 10 Additional Representative Peptides of the Invention Table 11 Immunogenicity of Representative Peptides of the Invention LIST OF SEQUENCES (1) GENERAL INFORMATION: (i) APPLICANT: UNITED BIOMEDICAL INC., Et al. (ii) TITLE OF THE INVENTION: PEPTIDAL COMPOSITION FOR THE PREVENTION AND TREATMENT OF HIV INFECTION AND IMMUNE DISEASES (iii) NUMBER OF SEQUENCES: 61 (iv) DIRECT CORRESPONDENCE: (A) RECIPIENT: Morgan & Finnegan, L.L.P. (B) STREET: 345 Park Avenue (C) CITY: New York (D) STATE: NY (E) COUNTRY: USA (F) POSTAL CODE: 10154-0054 (v) READING FORM ON THE COMPUTER: (A) TYPE MEDIUM: Flexible disk (B) COMPUTER: compatible with an IBM PC (C) OPERATING SYSTEM: PC-DOS / MS-DOS (D) SOFTWARE: Word 7.0 (vi) DATA FROM THE PREVIOUS REQUEST: (A) NO APPLICATION : US 09 / 100,409 (B) SUBMISSION DATE: June 20, 1998 (C) CLASSIFICATION: 514 (vii) CURRENT APPLICATION DATA: (A) APPLICATION NUMBER: To be determined (B) DATE OF SUBMISSION: 21 June 1999 (C) CLASSIFICATION: (viii) INFORMATION OF THE APPORTER / AGENT: (A) NAME: MARÍA CH LIN, ESQ. (B) REGISTRATION NUMBER: 29,323 (C) REFERENCE / FILE NUMBER: 1151-4154PC1 (ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 212-758-4800 (B) TELEFAX: 212-751-6849 (2) ) INFORMATION FOR SEC. IDENT. NO: 1: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 433 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (ix) DESCRIPTION OF THE SECIENCE: SEC. IDENT.:1: Lys Lys Val Val Leu Gly Lys Lys Gly Asp Thr Val 1 5 10 Giu Leu Thr Cys Thr Ala Ser Gln Lys Lys Ser lie 15 20 Gln Phe His Trp Lys Asn Ser Asn Gln lie Lys lie 25 30 35 Leu Gly Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro 40 45 Ser Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser 50 55 60 Leu Trp Asp Gln Gly Asn Phe Pro Leu Lie Lys 65 70 Asn Leu Lys lie Glu Asp Ser Asp Thr Tyr lie Cys 75 80 Glu Val Glu Asp Gln Lys Glu Glu Val Gln Leu Leu 85 90 95 Val Phe Gly Leu Thr Ala Asn Being Asp Thr His Leu 100 105 Leu Gln Gly Gln Ser Leu Thr Leu Thr Leu Glu Ser 110 115 120 Pro Pro Gly Ser Ser Pro Ser Val Gln Cys Arg Ser 125 130 Pro Arg Gly Lys Asn lie Gln Gly Gly Lys Thr Leu 135 140 Ser Val Ser Gln Leu Glu Leu Gln Asp Ser Gly Thr 145 150 155 Trp Thr Cys Thr Val Leu Gln Asn Gln Lys Lys Val 160 165 Glu Phe Lys lie Asp lie Val Val Leu Wing Phe Gln 170 175 180 Lys Wing Being Ser lie Val Tyr Lys Lys Glu Gly Glu 185 190 Gln Val Glu Phe Ser Phe Pro Leu Wing Phe Thr Val 195 200 Glu Lys Leu Thr Gly Ser Gly Glu Leu Trp Trp Gln 205 210 215 Ala Glu Arg Ala Ser Ser Ser Lys Ser Trp lie Me 220 225 Phe Asp Leu Lys Asn Lys Glu Val Ser Val Lys Arg 230 235 240 Val Thr Gln Asp Pro Lys Leu Gln Met Gly Lys Lys 245 250 Leu Pro Leu His Leu Thr Leu Pro Gln Ala Leu Pro 255 260 Gln Tyr Ala Gly Ser Gly Asn Leu Thr Leu Ala Leu 265 270 275 Glu Ala Lys Thr Gly Lys Leu His Gln Glu Val Asn 280 285 Leu Val Val Met Arg Ala Thr Gln Leu Gln Lys Asn 290 295 300 Leu Thr Cys Glu Val Trp Gly Pro Thr Ser Pro Lys 305 310 Leu Met Leu Ser Leu Lys Leu Glu Asn Lys Glu Ala 315 320 Lys Val Ser Lys Arg Glu Lys Pro Val Trp Val Leu 325 330 335 Asn Pro Glu Ala Gly Met Trp Gln Cys Leu Leu Ser 340 345 Asp Ser Ser Gln Val Leu Leu Glu Ser Asn Me Lys 350 355 360 Val Leu Pro Thr Trp Ser Thr Pro Val Gln Pro Met 365 370 Wing Leu Me Val Leu Gly Gly Val Wing Gly Leu Leu 375 380 Leu Phe Me Gly Leu Gly Me Phe Phe Cys Val Arg 385 390 395 Cys Arg His Arg Arg Arg Gln Wing Glu Arg Met Ser 400 405 Gln Me Lys Arg Leu Leu Ser Glu Lys Lys Thr Cys 410 415 420 Gln Cys Pro His Arg Phe Gln Lys Thr Cys Ser Pro 425 430 He (2) INFORMATION FOR SEC. IDENT. NO: 2: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 40 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 2: His Trp Lys Asn Trp Asn Gln Me Lys Me Leu Gly 1 5 10 Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 15 20 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 25 30 35 Asp Gln Gly Asn 40 (2) INFORMATION FOR SEC. IDENT. NO: 3: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 28 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 3: Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 1 5 10 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 15 20 Asp Gln Gly Asn 25 (2) INFORMATION FOR SEC. IDENT. NO: 4: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 42 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 4: Cys His Trp Lys Asn Trp Asn Gln Me Lys Me Leu 1 5 10 Gly Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser 15 20 Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu 25 30 35 Trp Asp Gln Gly Asn Cys 40 (2) INFORMATION FOR SEC. IDENT. NO: 5: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 30 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 5: Cys Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser 1 5 10 Lys Leu Asn Asp Arg Ala. Asp Ser Arg Arg Ser Leu 15 20 Trp Asp Gln Gly Asn Cys 25 30 (2) INFORMATION FOR SEC. IDENT. NO: 6: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 15 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (ix) CHARACTERISTIC: (A) NAME / KEY: Modified site (B) LOCATION: 1 (D) OTHER INFORMATION: / note = "Me, Met or Leu" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 2 (D) OTHER INFORMATION: / note = "Ser or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) ) LOCATION: 4 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 5 (D) OTHER INFORMATION: / note = "Gly or Thr "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 10 (D) OTHER INFORMATION: / note =" His or Thr "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 11 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 12 (D) OTHER INFORMATION: / note = " Me, Met or Leu "(¡x) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 14 (D) OTHER INFORMATION: / note =" Gly or Thr "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 15 (D) OTHER INFORMATION: / note = "Me, Met or Val" (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 6: Xaa Xaa Glu Xaa Xaa Gly Val Me Val Xaa Xaa Xaa 1 5 10 Glu Xaa Xaa 15 (2) INFORMATION FOR SEC. IDENT. NO: 7: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 16 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 7: Thr Ala Lys Ser Lys Lys Phe Pro Ser Tyr Thr Ala 1 5 10 Thr Tyr Gln Phe 15 (2) INFORMATION FOR SEC. IDENT. NO: 8: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 15 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 8: Phe Phe Leu Leu Thr Arg Me Leu Thr Me Pro Gln 1 5 10 Ser Leu Asp 15 (2) INFORMATION FOR SEC. IDENT. NO: 9: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 6 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 9: Pro Pro Xaa Pro Xaa Pro 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 10: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 46 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 10: Cys His Trp Lys Asn Trp Asn Gln Me Lys Me Leu 1 5 10 Gly Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser 15 20 Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu 25 30 35 Trp Asp Gln Gly Asn Cys Pro Leu Me Me 40 45 (2) INFORMATION FOR SEC. IDENT. NO: 11: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 34 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 11: Cys Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser 1 5 10 Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu 15 20 Trp Asp Gln Gly Asn Cys Pro Leu e Me 25 30 (2) INFORMATION FOR SEC. IDENT. NO: 12: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 18 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 12: Lys Lys Lys Me Me Thr Me Thr Arg Me Me Thr 1 5 10 Me Me Thr Thr Me Asp 15 (2) INFORMATION FOR SEC. IDENT. NO: 13: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 12 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 13: Thr Me Asn Lys Pro Lys Gly Tyr Val Gly Lys Glu 1 5 10 (2) INFORMATION FOR SEC. IDENT. NO: 14: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 13 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 14: Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp Arg Ala 1 5 10 (2) INFORMATION FOR SEC. IDENT. NO: 15: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 12 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 15: Cys Gly Asn Gln Gly Ser Phe Leu Thr Lys Gly Cys 1 5 10 (2) INFORMATION FOR SEC. IDENT. NO: 16: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 18 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 16: Gly Pro Ser Lys Leu Asn Asp Arg Wing Asp Ser Arg 1 5 10 Arg Ser Leu Trp Asp Gln 15 (2) INFORMATION FOR SEC. IDENT. NO: 17: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 7 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 17: Asn Gln Gly Ser Phe Leu Thr 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 18: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 12 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 18: Cys Me Leu Gly Asn Gln Gly Ser Phe Leu Thr Cys 1 5 10 (2) INFORMATION FOR SEC. IDENT. NO: 19: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 29 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 19: Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 1 5 10 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 15 20 Asp Gln Gly Asn Phe 25 (2) INFORMATION FOR SEC. IDENT. NO: 20: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 61 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 20: Ser Lys Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser 1 5 10 Leu Trp Asp Gln Gly Asn Phe Pro Leu Me Me Lys 15 20 Asn Leu Lys Me Glu Asp Ser Asp Thr Tyr Me Cys 25 30 35 Glu Val Glu Asp Gln Lys Glu Glu Val Gln Leu Leu 40 45 Val Phe Gly Leu Thr Wing Asn Ser Asp Thr His Leu 50 55 60 Leu (2) INFORMATION FOR SEC. IDENT. NO: 21: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 14 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 21: Cys Me Leu Gly Asn Gln Gly Ser Phe Leu Thr Lys 1 5 10 Gly Cys (2) INFORMATION FOR SEC. IDENT. NO: 22: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 22: Cys Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn 1 5 10 Asp Arg Wing Asp Ser Arg Arg Cys 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 23: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 15 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 23: Cys Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp Arg 1 5 10 Wing Asp Cys 15 (2) INFORMATION FOR SEC. IDENT. NO: 24: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 12 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 24: Cys Lys Gly Pro Ser Lys Leu Asn Asp Arg Ala Cys 1 5 10 (2) INFORMATION FOR SEC. IDENT. NO: 25: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 25: Cys Ser Asp Thr Tyr Me Cys Glu Val Glu Asp Gln 1 5 10 Lys Glu Glu Val Gln Leu Leu Cys 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 26: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 10 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 26: Cys Gly Asn Gln Gly Ser Phe Leu Thr Cys 1 5 10 (2) INFORMATION FOR SEC. IDENT. NO: 27: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 8 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 27: Cys Asn Gln Gly Ser Phe Leu Cys 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 28: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 6 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 28: Cys Gln Gly Ser Phe Cys 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 29: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 5 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 29: Cys Asn Thr Arg Cys 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 30: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 7 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 30: Cys Leu Asn Thr Arg Ala Cys 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 31: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 7 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 31: Cys Pro Ser Lys Leu Asn Cys 1 5 (2) INFORMATION FOR SEC. IDENT. NO: 32: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 77 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 32: Thr Ala Lys Ser Lys Lys Phe Pro Ser Tyr Thr Ala 1 5 10 Thr Tyr Gln Phe Gly Gly Phe Phe Leu Leu Thr Arg 15 20 Me Leu Thr Me Pro Gln Ser Leu Asp Gly Gly Cys 25 30 35 His Trp Lys Asn Trp Asn Gln Me Lys Me Leu Gly 40 45 Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 50 55 60 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 65 70 Asp Gln Gly Asn Cys 75 (2) INFORMATION FOR SEC . IDENT. NO: 33: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 69 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 33: Thr Ala Lys Ser Lys Lys Phe Pro Ser Tyr Thr Ala 1 5 10 Thr Tyr Gln Phe Gly Gly Phe Phe Leu Thu Arg 15 20 Me Leu Thr Me Pro Gln Ser Leu Asp Gly Gly Cys 25 30 35 Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 40 45 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 50 55 60 Asp Gln Giy Asn Cys Pro Leu Me Me 65 (2) INFORMATION FOR SEC. IDENT. NO: 34: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 81 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 34: Thr Ala Lys Ser Lys Lys Phe Pro Ser Tyr Thr Ala 1 5 10 Thr Tyr Gln Phe Gly Gly Phe Phe Leu Leu Thr Arg 15 20 Me Leu Thr Me Pro Gln Ser Leu Asp Gly Gly Cys 25 30 35 His Trp Lys Asn Trn Asn Gln Me Lys Me Leu Gly 40 45 Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 50 55 60 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 65 70 Asp Gln Gly Asn Cys Pro Leu Me Me 75 80 (2) INFORMATION FOR SEC. IDENT. NO: 35: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 64 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 35: Thr Ala Lys Ser Lys Lys Phe Pro Ser Tyr Thr Ala 1 5 10 Thr Tyr Gln Phe Gly Gly Phe Phe Leu Leu Thr Arg 15 20 Me Leu Thr Me Pro Gln Ser Leu Asp Gly Gly Cys 25 30 35 Asn Gln Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys 40 45 Leu Asn Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp 50 55 60 Asp Gln Gly Cys (2) INFORMATION FOR SEC. IDENT. NO: 36: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 19 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (ix) CHARACTERISTICS: (A) NAME / KEY : Modified site (B) LOCATION: 4 (D) OTHER INFORMATION: / note = "Ser or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 7 (D) OTHER INFORMATION: / note = " Lys or Arg "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 8 (D) OTHER INFORMATION: / note =" Gly or Thr "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 12 (D) OTHER INFORMATION: / note = "His or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 13 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 16 (D) OTHER INFORMATION: / note = "Gly or Thr" (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 36: Me Ser Xaa Glu Me Xaa Xaa Val Me Val Xaa 1 5 10 Xaa Me Glu Xaa Me Leu Phe 15 (2) INFORMATION FOR SEC. IDENT. NO: 37: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 50 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 37: Lys Lys Me Thr Me Thr Arg Me Me Thr 1 5 10 lie Me Thr Thr Me Asp Gly Gly Cys Asn Gln Gly 15 20 Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn Asp 25 30 35 Arg Ala Asp Ser Arg Arg Ser Leu Trp Asp Gln Gly 40 45 Asn Cys 50 (2) INFORMATION FOR SEC. IDENT. NO: 38: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 15 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 38: Leu Ser Glu Me Lys Gly Val Me Val His Arg Leu 1 5 10 Glu Gly Val 15 (2) INFORMATION FOR SEC. IDENT. NO: 39: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 39: Gly Me Leu Glu Ser Arg Gly Me Lys Ala Arg Me 1 5 10 Thr His Val Asp Thr Glu Ser Tyr 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 40: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 17 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 40: Lys Lys Gln Tyr Me Lys Wing Asn Ser Lys Phe Me 1 5 10 Gly Me Thr Glu Leu 15 (2) INFORMATION FOR SEC. IDENT. NO: 41: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 22 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 41: Lys Lys Phe Asn Asn Phe Thr Val Ser Phe Trp Leu 1 5 10 Arg Val Pro Lys Val Ser Wing Ser His Leu 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 42: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 30 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 42: Lys Lys Leu Arg Arg Leu Leu Tyr Met Me Tyr Met 1 5 10 Ser Gly Leu Ala Val Arg Val His Val Ser Lys Glu 15 20 Glu Gln Tyr Tyr Asp Tyr 25 30 (2) INFORMATION FOR SEC. IDENT. NO: 43: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 27 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 43: Tyr Asp Pro Asn Tyr Leu Arg Thr Asp Ser Asp Lys 1 5 10 Asp Arg Phe Leu Gln Thr Met Val Lys Leu Phe Asn 15 20 Arg Me Lys 25 (2) INFORMATION FOR SEC. IDENT. NO: 44: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 24 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 44: Gly Ala Tyr Ala Arg Cys Pro Asn Gly Thr Arg Ala 1 5 10 Leu Thr Val Ala Glu Leu Arg Gly Asn Ala Glu Leu 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 45: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 21 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 45: Val Ser Phe Gly Val Trp Me Arg Thr Pro Pro Wing 1 5 10 Tyr Arg Pro Pro Asn Ala Pro Me Leu 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 46: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 46: Ser Asp Phe Phe Pro Ser Val Arg Asp Leu Leu Asp 1 5 10 Thr Ala Ser Ala Leu Tyr Arg Glu 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 47: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 47: Pro His His Thr Wing Leu Arg Gln Wing Me Leu Cys 1 5 10 Trp Gly Glu Leu Met Thr Leu Wing 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 48: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 17 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 48: Trp Val Arg Asp Me Me Asp Asp Phe Thr Asn Glu 1 5 10 Be Ser Gln Lys Thr 15 (2) INFORMATION FOR SEC. IDENT. NO: 49: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 19 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 49: Arg Ala Gly Arg Ala Me Leu His Me Pro Thr Arg 1 5 10 Me Arg Gln Gly Leu Glu Arg 15 (2) INFORMATION FOR SEC. IDENT. NO: 50: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 21 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 50: Wing Val Wing Glu Gly Thr Asp Arg Val Me Glu Val 1 5 10 Leu Gln Arg Wing Gly Arg Wing Me Leu 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 51: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 25 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 51: Wing Leu Asn Me Trp Asp Arg Phe Asp Val Phe Ser 1 5 10 Thr Leu Gly Wing Thr Ser Gly Tyr Leu Lys Gly Asn 15 20 Ser 25 (2) INFORMATION FOR SEC. IDENT. NO: 52: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 22 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 52: Asp Ser Glu Thr Wing Asp Asn Leu Glu Lys Thr Val 1 5 10 Ala Ala Leu Ser Me Leu Pro Gly His Gly 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 53: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 39 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 53: Glu Glu Me Val Wing Gln Being Me Wing Leu Being 1 5 10 Leu Met Val Wing Gln Wing Me Pro Leu Val Gly Glu 15 20 Leu Val Asp Me Gly Phe Wing Wing Thr Asn Phe Val 25 30 35 Glu Ser Cys (2) INFORMATION FOR SEC. IDENT. NO: 54: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 21 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 54: Asp Me Glu Lys Lys Me Wing Lys Met Glu Lys Wing 1 5 10 Ser Ser Val Phe Asn Val Val Asn Ser 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 55: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 17 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 55: Lys Trp Phe Lys Thr Asn Ala Pro Asn Gly Val Asp 1 5 10 Glu Lys Me Arg Me 15 (2) INFORMATION FOR SEC. IDENT. NO: 56: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 14 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 56: Gly Leu Gln Gly Lys Me Wing Asp Wing Val Lys Wing 1 5 10 Lys Gly (2) INFORMATION FOR SEC. IDENT. NO: 57: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 19 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 57: Gly Leu Wing Wing Gly Leu Val Gly Met Wing Wing Asp 1 5 10 Wing Met Val Glu Asp Val Asn 15 (2) INFORMATION FOR SEC. IDENT. NO: 58: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (xi) DESCRIPTION OF SEQUENCE: SEC. IDENT. NO: 58: Ser Thr Glu Thr Gly Asn Gln His His Tyr Gln Thr 1 5 10 Arg Val Val Ser Asn Wing Asn Lys 15 20 (2) INFORMATION FOR SEC. IDENT. NO: 59: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 19 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (ix) CHARACTERISTIC: (A) NAME / KEY : Modified site (B) LOCATION: 3 (D) OTHER INFORMATION: / note = "Me, Met or Leu" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 4 (D) OTHER INFORMATION : / note = "Ser or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 7 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 8 (D) OTHER INFORMATION: / note = " Gly or Thr "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 12 (D) OTHER INFORMATION: / note =" His or Thr "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 13 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 14 (D) OTHER INFORMATION: / note = "Me, Met or Leu "(ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 16 (D) OTHER INFORMATION: / note =" Gly or Thr "(ix) FEATURE: (A) NAME / KEY : Modified site (B) LOCATION: 17 (D) OTHER INFORMATION: / note = "Me, Met or Val" (xi) DESCRIPTION OF THE SEQUENCE: SECTION ID: 59: Me Be Xaa Xaa Glu Me Xaa Xaa Val Me Val Xaa 1 5 10 Xaa Xaa Glu Xaa Xaa Leu Phe 15 (2) INFORMATION FOR SECTION ID NO: 60: (i) CHARACTERISTICS OF THE SEQUENCE: (A) LENGTH: 51 amino acids (B) TYPE: amino acid ( D) TOPOLOGY: linear (i) TYPE OF MOLECULE: peptide (ix) CHARACTERISTIC: (A) NAME / KEY: Modified site (B) LOCATION: 4 (D) OTHER INFORMATION: / note = "Ser or Thr" (¡) x) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 7 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 8 (D) OTHER INFORMATION: / note = "Gly or Thr" (¡x) CHARACTERISTICS: (A) NAME / CLA VE: Modified site (B) LOCATION: 12 (D) OTHER INFORMATION: / note = "His or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 13 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 16 (D) OTHER INFORMATION: / note = "Gly or Thr" (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 60: Me Being Xaa Glu Me Xaa Xaa Val Me Val Xaa 1 5 10 Xaa Me Glu Xaa Me Leu Phe Gly Gly Cys Asn Gln 15 20 Gly Ser Phe Leu Thr Lys Gly Pro Ser Lys Leu Asn 25 30 35 Asp Arg Wing Asp Ser Arg Arg Ser Leu Trp Asp Gln 40 45 Gly Asn Cys 50 (2) INFORMATION FOR SEC. IDENT. NO: 61: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 70 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) TYPE OF MOLECULE: peptide (ix) CHARACTERISTICS: (A) NAME / KEY : Modified site (B) LOCATION: 24 (D) OTHER INFORMATION: / note = "Me, Met or Leu" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 25 (D) OTHER INFORMATION : / note = "Ser or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 27 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) CHARACTERISTICS: (A) ) NAME / KEY: Modified site (B) LOCATION: 28 (D) OTHER INFORMATION: / note = "Gly or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 33 (D) OTHER INFORMATION: / note = "His or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 34 (D) OTHER INFORMATION: / note = "Lys or Arg" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 35 (D) OTHER INFORMATION: / note = "Me, Met or Leu" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 37 (D) OTHER INFORMATION: / note = "Gly or Thr" (ix) FEATURE: (A) NAME / KEY: Modified site (B) LOCATION: 38 (D) OTHER INFORMATION: / note = "Me, Met or Val" (xi) DESCRIPTION OF THE SEQUENCE: SEC. IDENT. NO: 61 Wing Val Wing Glu Gly Thr Asp Arg Val Me Glu Val 1 5 10 Leu Gln Arg Ala Gly Arg Ala Me Leu Gly Gly Xaa 15 20 Xaa Glu Xaa Xaa Gly Val Me Val Xaa Xaa Xaa Glu 30 35 Xaa Xaa Gly Gly Cys Asn Gln Gly Ser Phe Leu Thr 40 45 Lys Gly Pro Ser Lys Leu Asn Asp Arg Wing Asp Ser 50 55 60 Arg Arg Ser Leu Trp Asp Gln Gly Asn Cys 65 70

Claims (25)

1. A CD4-CDR2 antigen peptide, characterized in that the antigen peptide is between about 30 and about 46 amino acids in length; wherein the peptide antigen CD4-CDR2 contains two cysteine residues separated by an interposed sequence of 28 to 40 amino acid residues; and wherein the intervening sequence is a contiguous portion of the sequence represented by residues 27 to 66 of SEC. FROM IDENT. NO: 1, or is an immunologically functional homologue of residues 27 to 66 of SEC. FROM IDENT. NO: 1.
2. The CD4-CDR2 antigen peptide of claim 1, characterized in that the antigen peptide is selected from the group consisting of SEC. FROM IDENT. NO: 4, SEC. FROM IDENT. NO: 5, SEC. FROM IDENT. NO: 10, SEC. FROM IDENT. NO: 11, and immunologically functional homologs thereof.
3. The synthetic peptide of about 50 to about 80 amino acids in length, characterized in that it comprises (a) an attendant T-cell epitope (Th), (b) a CD4-CDR2 antigen peptide according to claim 1; and (c) an immunostimulatory invasin domain.
4. The synthetic peptide of about 50 to about 80 amino acids in length characterized in that it comprises (a) an attendant T cell epitope (Th), (b) a CD4-CDR2 antigen peptide according to claim 2; and (c) an immunostimulatory invasin domain.
5. The peptide or peptide conjugate characterized in that it comprises an attendant T cell epitope (Th) covalently linked to a CD4-CDR2 antigen peptide according to claim 1.
6. The peptide or peptide conjugate characterized in that it comprises epitope (Th) of assistant T-cell covalently bound to a CD4-CDR2 antigen peptide according to claim 2.
7. The peptide or peptide conjugate represented by the formula (A) n- (CD4-CDR2 peptide antigen) - (B) 0- ( Th) mX or (A) n- (Th) m- (B) 0- (CD4-CDR2 peptide antigen) -X characterized in that each A is independently an amino acid or a general immunostimulatory sequence; each B is selected from the group consisting of amino acids, -NHCH (X) CH2SCH2CO-, -NHCH (X) CH2SCH2CO (e- N) Lys-, -NHCH (X) CH2S-succinimidyl (eN) Lys-, and -NHCH (X) CH2S- (succinimidyl) -; each Th is independently a sequence of amino acids comprising an attendant T-cell epitope, or an immunomodulatory analog or segments thereof; the CD4-CDR2 antigen peptide represents the sequence of an antigen peptide according to claim 1; X is an amino acid a-COOH or a-CONH2; n is from 0 to about 10; m is from 1 to about 4; I is from 0 to about 10.
8. The peptide or peptide conjugate represented by the formula (CD4-CDR2 peptide antigen) - (B) 0- (Th) m- (A) nX (Th) m- (B) 0 - (CD4-CDR2 peptide antigen) - (A) nX characterized in that each A is independently an amino acid or a general immunostimulatory sequence; each B is selected from the group consisting of amino acids, -NHCH (X) CH2SCH2CO-, -NHCH (X) CH2SCH2CO (eN) Lys-, -NHCH (X) CH2S-succinimidyl (eN) Lys-, and -NHCH (X) CH2S- (succinimidyl) -; each Th is independently an amino acid sequence comprising an attendant T cell epitope, or an immunomodulatory analog or segment thereof; the CD4-CDR2 antigen peptide represents the sequence of an antigen peptide according to claim 1; X is an amino acid a-COOH or a-CONH2; n is from 0 to about 10; m is from 1 to about 4; I is from 0 to about 10.
9. The peptide or peptide conjugate represented by the formula (A) n- (CD4-CDR2 peptide antigen) - (B) 0- (Th) mX or (A) n- (Th) m- (B) 0- (CD4-CDR2 peptide antigen) -X characterized in that each A is independently an amino acid or a general immunostimulatory sequence; each B is selected from the group consisting of amino acids, -NHCH (X) CH2SCH2CO-, -NHCH (X) CH2SCH2CO (eN) Lys-, -NHCH (X) CH2S-succinimidyl (eN) Lys-, and -NHCH (X) CH2S- (succinimidyl) -; each Th is independently a sequence of amino acids comprising an attendant T cell epitope, or an immunomodulatory analog or segment thereof; the peptide antigen CD4-CDR2 represents the sequence of an antigen peptide according to claim 2; X is an amino acid a-COOH or a-CONH2; n is from 0 to about 10; m is from 1 to about 4; I is from 0 to about
10. 10. The peptide or peptide conjugate represented by the formula (CD4-CDR2 peptide antigen) - (B) 0- (Th) m- (A) nX or (Th) m- (B) 0- (CD4-CDR2 peptide antigen) - (A) nX characterized in that each A is independently an amino acid or a general immunostimulatory sequence; each B is selected from the group consisting of amino acids, -NHCH (X) CH2SCH2CO-, -NHCH (X) CH2SCH2CO (eN) Lys-, -NHCH (X) CH2S-succinimidyl (eN) Lys-, and -NHCH (X) CH2S- (succinimidyl) -; each Th is independently a sequence of amino acids comprising an attendant T-cell epitope, or an enhancer analog or segment thereof; the peptide antigen CD4-CDR2 represents the sequence of an antigen peptide according to claim 2; X is an amino acid a-COOH or a-CONH2; n is from 0 to about 10; m is from 1 to about 4; and o is from 0 to about 10. The peptide or peptide conjugate according to any of claims 3-10, characterized in that the Th has an amino acid sequence selected from the group consisting of SEC. FROM IDENT. US: 6, 8, 12, 13, 36, and 38-59. 12. The peptide or peptide conjugate according to claim 11, characterized in that the Th has an amino acid sequence selected from the group consisting of SEC. FROM IDENT. NO: 6 and SEC. FROM IDENT. NO: 8 13. The peptide or peptide conjugate according to any of claims 3-10, characterized in that n is 3, and (A) 3 is (invasin domain) -Gly-Gly. 14. The peptide or peptide conjugate according to claim 11, characterized in that at least one portion A is an invasin domain. 15. The peptide or peptide conjugate according to claim 12, characterized in that at least one portion A is an invasin domain. 16. The peptide or peptide conjugate according to any of claims 3-10, characterized in that the peptide antigen CD4-CDR2 is selected from the group consisting of SEC. FROM IDENT. NOS: 4, 5, 10, and
11. 17. The peptide or peptide conjugate according to claim 11, characterized in that the peptide antigen CD4-CDR2 is selected from the group consisting of SEC. FROM IDENT. NOS: 4, 5, 10, and 11. 18. The peptide or peptide conjugate according to claim 12, characterized in that the peptide antigen CD4-CDR2 is selected from the group consisting of SEC. FROM IDENT. NOS: 4, 5, 10, and 11. 19. A peptide selected from the group consisting of SEC. FROM IDENT. NOS: 32, 33, 34, 35, and 60. 20. A pharmaceutical composition comprising an immunologically effective amount of a peptide or peptide conjugate according to any of claims 3-10 or 19, further characterized in that it additionally comprises a carrier. pharmaceutically acceptable. 21. The pharmaceutical composition according to claim 20, characterized in that the immunologically effective amount of the peptide or peptide conjugate is between about 0.5 μg and about 1 mg per kilogram of body weight per dose. 22. The method for inducing antibodies to a CD4 surface complex in a mammal which is characterized in that it comprises administering to the mammal a pharmaceutical composition according to claim 20. 23. The method for inducing antibodies to the CD4 surface complex in a mammal which is characterized in that it comprises administering to the mammal a pharmaceutical composition according to claim 21. 24. The method for inhibiting the binding of HIV to CD4 + cells in a mammal which comprises administering to the mammal a pharmaceutical composition in accordance with claim 20. 25. The method for inhibiting the binding of HIV to CD4 + cells in a mammal which is characterized in that it comprises administering to the mammal a pharmaceutical composition according to claim 21.
MXPA/A/2000/011508A 1998-06-20 2000-11-22 Antigenic complex comprising immunostimulatory peptide, cd4, and chemokine receptor domain for hiv treatment and immune disorders MXPA00011508A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09100409 1998-06-20

Publications (1)

Publication Number Publication Date
MXPA00011508A true MXPA00011508A (en) 2002-03-26

Family

ID=

Similar Documents

Publication Publication Date Title
EP1098910B1 (en) Antigenic complex comprising immunostimulatory peptide, cd4, and chemokine receptor domain for hiv treatment and immune disorders
US6107021A (en) Synthetic peptide vaccines for foot-and-mouth disease
JP3802049B2 (en) Composite synthetic peptide constructs that induce neutralizing antibodies to HIV and cytotoxic T lymphocytes
AU732809B2 (en) Compositions and methods for treating viral infections
EP0328403A2 (en) Synthetic peptides related to the HIV-GP120-env-protein, and their use
US20100003281A1 (en) Monoclonal antibody against hepatitis e virus or its fragment with binding activity and use thereof
WO2000040616A1 (en) METHODS OF ELICITING BROADLY NEUTRALIZING ANTIBODIES TARGETING HIV-1 gp41
NZ230855A (en) Principal neutralising domain of hiv variant, dna, vaccine, assay, antibodies and treatment
Boudet et al. UV-treated polystyrene microtitre plates for use in an ELISA to measure antibodies against synthetic peptides
EP0492560B1 (en) Human monoclonal antibodies directed against the transmembrane glycoprotein (gp41) of HIV-1, and related peptides
JP4307838B2 (en) Peptide showing affinity for GP120 viral protein and use thereof
CA2160696C (en) Peptides for use in vaccination and induction of neutralizing antibodies against human immunodeficiency virus
MXPA00011508A (en) Antigenic complex comprising immunostimulatory peptide, cd4, and chemokine receptor domain for hiv treatment and immune disorders
AU2006200454B2 (en) Compositions and methods for treating viral infections
WO1989010939A1 (en) Immunogens and biologically active peptides derived from shared sequences from antigens and anti-idiotypic antibodies or antibodies specific for cellular receptors of the antigens
AU2008203501A1 (en) Compositions and methods for treating viral infections
MXPA99003380A (en) Compositions and methods for treating viral infections
AU2006200455A1 (en) Compositions and methods for treating viral infections