Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTIGEN-ANTIBODY COMPLEXES AS HIV-1 VACCINES
Document Type and Number:
WIPO Patent Application WO/2008/127391
Kind Code:
A3
Abstract:
The present relation relates to antigen-antibody complexes for use as prophylactic and therapeutic vaccines for infectious diseases of AIDS. The present invention encompasses the preparation and purification of immunogenic antibody-antigen complexes which formulated into the vaccines of the present invention.

Inventors:
KOFF WAYNE (US)
Application Number:
PCT/US2007/083006
Publication Date:
September 03, 2009
Filing Date:
October 30, 2007
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INT AIDS VACCINE INITIATIVE (US)
International Classes:
A61K39/00
Foreign References:
US20020086022A12002-07-04
US20060051373A12006-03-09
US20050124540A12005-06-09
Attorney, Agent or Firm:
KOWALSKI, Thomas, J. et al. (745 Fifth AvenueNew York, NY, US)
Download PDF:
Claims:

WHAT IS CLAIMED IS:

1. A method of producing an immune response comprising administering to a mammal a purified antibody-antigen complex dissociated from polyclonal anti-HIV sera bound to glycoprotein spikes on HIV envelopes.

2. A method of producing an immune response comprising administering to a mammal a purified antibody-antigen complex dissociated from a mixture of broadly neutralizing antibodies and HIV, wherein the mixture is bound to glycoprotein spikes on HIV envelopes.

3. The method of claim 2 wherein the antibodies are monoclonal antibodies.

4. The method of claim 3 wherein the monoclonal antibodies are bl2, 2F5, 2Gl 2, 4E10, M2909 or any combination thereof.

5. The method of claim 2 wherein the HIV is purified HIV.

6. The method of claim 2 wherein the HIV is a HIV viral isolate.

7. The method of claim 6 wherein the HIV viral isolate is a HIV clade viral isolate.

8. The method of claim 1 or 2 wherein the purified antibody-antigen complex is chemically dissociated from the glycoprotein spikes.

9. The method of claim 1 or 2 wherein the purified antibody-antigen complex is dissociated from the glycoprotein spikes by solubilizing a HIV lipid bilayer.

10. The method of claim 1 or 2 wherein the purified antibody-antigen complex is purified with Protein A, protein G, precipitating secondary antibodies or Protein A-bearing S. aureus cells.

11. The method of claim 1 or 2 wherein the mammal is a human.

12. The method of claim 1 or 2 wherein the purified antibody-antigen complex is administered in a pharmaceutically acceptable carrier.

13. The method of claim 1 or 2 wherein the administering further comprises a prime-boost regimen.

Description:

TITLE OF THE INVENTION

Antigen - Antibody Complexes as HIV-I Vaccines INCORPORATION BY REFERENCE

This application claims priority to United States Provisional Application No. 60/855,625, filed on 30 October 2006.

The foregoing applications, and all documents cited therein or during their prosecution ("appln cited documents") and all documents cited or referenced in the appln cited documents, and all documents cited or referenced herein ("herein cited documents"), and all documents cited or referenced in herein cited documents, together with any manufacturer's instructions, descriptions, product specifications, and product sheets for any products mentioned herein or in any document incorporated by reference herein, are hereby incorporated herein by reference, and may be employed in the practice of the invention. FIELD OF THE INVENTION

The present invention relates to antigen-antibody complexes for use as prophylactic and therapeutic vaccines for infectious diseases of AIDS. BACKGROUND OF THE INVENTION

AIDS, or Acquired Immunodeficiency Syndrome, is caused by human immunodeficiency virus (HIV) and is characterized by several clinical features including wasting syndromes, central nervous system degeneration and profound immunosuppression that results in opportunistic infections and malignancies. HIV is a member of the lentivirus family of animal retroviruses, which include the visna virus of sheep and the bovine, feline, and simian immunodeficiency viruses (SIV). Two closely related types of HIV, designated HIV-I and HIV -2, have been identified thus far, of which HIV-I is by far the most common cause of AIDS. However, HIV-2, which differs in genomic structure and antigenicity, causes a similar clinical syndrome.

An infectious HIV particle consists of two identical strands of RNA, each approximately 9.2 kb long, packaged within a core of viral proteins. This core structure is surrounded by a phospholipid bilayer envelope derived from the host cell membrane that also includes virally-encoded membrane proteins (Abbas et al., Cellular and Molecular Immunology, 4th edition, W.B. Saunders Company, 2000, p. 454). The HIV genome has the characteristic 5'-LTR-Gag-Pol-Env-LTR-3' organization of the retrovirus family. Long terminal repeats (LTRs) at each end of the viral genome serve as binding sites for transcriptional regulatory proteins from the host and regulate viral integration into the host genome, viral gene expression, and viral replication.

The HIV genome encodes several structural proteins. The Gag gene encodes core structural proteins of the nucleocapsid core and matrix. The Pol gene encodes reverse transcriptase (RT), integrase (Int), and viral protease enzymes required for viral replication. The tat gene encodes a protein that is required for elongation of viral transcripts. The rev gene encodes a protein that promotes the nuclear export of incompletely spliced or unspliced viral RNAs. The Vif gene product enhances the infectivity of viral particles. The vpr gene product promotes the nuclear import of viral DNA and regulates G2 cell cycle arrest. The vpu and nef genes encode proteins that down regulate host cell CD4 expression and enhance release of virus from infected cells. The Env gene encodes the viral envelope glycoprotein that is translated as a 160-kilodalton (kDa) precursor (gpl60) and cleaved by a cellular protease to yield the external 120-kDa envelope glycoprotein (gpl20) and the transmembrane 41 -kDa envelope glycoprotein (gp41), which are required for the infection of cells (Abbas, pp. 454-456). Gpl40 is a modified form of the env glycoprotein which contains the external 120-kDa envelope glycoprotein portion and a part of the gp41 portion of env and has characteristics of both gp 120 and gp41. The Nef gene is conserved among primate lentiviruses and is one of the first viral genes that is transcribed following infection. In vitro, several functions have been described, including down regulation of CD4 and MHC class I surface expression, altered T-cell signaling and activation, and enhanced viral infectivity. HIV infection initiates with gpl20 on the viral particle binding to the CD4 and chemokine receptor molecules (e.g., CXCR4, CCR5) on the cell membrane of target cells such as CD4+ T-cells, macrophages and dendritic cells. The bound virus fuses with the target cell and reverse transcribes the RNA genome. The resulting viral DNA integrates into the cellular genome, where it directs the production of new viral RNA, and thereby viral proteins and new virions. These virions bud from the infected cell membrane and establish productive infections in other cells. This process also kills the originally infected cell. HIV can also kill cells indirectly because the CD4 receptor on uninfected T-cells has a strong affinity for gpl20 expressed on the surface of infected cells. In this case, the uninfected cells bind, via the CD4 receptor-gpl20 interaction, to infected cells and fuse to form a syncytium, which cannot survive. Destruction of CD4+ T-lymphocytes, which are critical to immune defense, is a major cause of the progressive immune dysfunction that is the hallmark of AIDS disease progression. The loss of CD4+ T cells seriously impairs the body's ability to fight most invaders, but it has a particularly severe impact on the defenses against viruses, fungi, parasites and certain bacteria, including mycobacteria.

Research on the Env glycoproteins have shown that the virus has many effective protective mechanisms with few vulnerabilities (Wyatt & Sodroski, Science. 1998 Jun 19;280(5371):1884-8). For fusion with its target cells, HIV-I uses a trimeric Env complex containing gpl20 and gp41 subunits (Burton et al., Nat Immunol. 2004 Mar;5(3):233-6). The fusion potential of the Env complex is triggered by engagement of the CD4 receptor and a coreceptor, usually CCR5 or CXCR4. Neutralizing antibodies seem to work either by binding to the mature trimer on the virion surface and preventing initial receptor engagement events or by binding after virion attachment and inhibiting the fusion process (Parren & Burton, Adv Immunol. 2001;77:195-262). In the latter case, neutralizing antibodies may bind to epitopes whose exposure is enhanced or triggered by receptor binding. However, given the potential antiviral effects of neutralizing antibodies, it is not unexpected that HIV-I has evolved multiple mechanisms to protect it from antibody binding (Johnson & Desrosiers, Annu Rev Med. 2002;53:499-518).

There remains a need to identify immunogens that elicit broadly neutralizing antibodies. Strategies include producing molecules that mimic the mature trimer on the virion surface, producing Env molecules engineered to better present neutralizing antibody epitopes than wild-type molecules, generating stable intermediates of the entry process to expose conserved epitopes to which antibodies could gain access during entry and producing epitope mimics of the broadly neutralizing monoclonal antibodies determined from structural studies of the antibody-antigen complexes (Burton et al., Nat Immunol. 2004 Mar;5(3):233- 6). However, none of these approaches have yet efficiently elicited neutralizing antibodies with broad specificity.

Citation or identification of any document in this application is not an admission that such document is available as prior art to the present application. SUMMARY OF THE INVENTION

The current invention is based, in part, on Applicant's discovery that immunization with antigen-antibody complexes elicit neutralizing antibody responses. Broadly neutralizing antibodies, if passively administered to monkeys, protect against an HIV equivalent virus (SIV/HIV chimera, e.g., SHIV). The identification of antigens that bind the neutralizing antibodies remains challenging, especially elucidating the preferred conformation of the antigen.

The solution proposed by the present invention is immunization with the antibody- antigen complex, wherein the antigen is held in its preferred conformation by the antibody or its equivalent polyclonal sera. One of skill in the art would not expect this approach to work

as the antigen are bound to the antibody and the epitopes are covered. Without being bound by theory, it is hypothesized that an antibody-antigen complex is presented to the immune system in a novel form, is dissociated within the antigen presenting cells and elicits the correct antibody response. The present invention encompasses identification of antibody-antigen complexes for use as a HIV vaccine. In one embodiment, the invention relates to the identification of immunogenic antibody-antigen complexes.

In one embodiment, mixing polyclonal anti-HIV sera which demonstrate broad neutralizing activity with purified HIV enables the antibodies to bind to the glycoprotein spikes on the viral envelopes. The antibody-antigen complexes are dissociated, advantageously chemically dissociated, from the virus. The antibody-antigen complexes are purified and formulated into the vaccines of the present invention.

In another embodiment, broadly neutralizing HIV monoclonal antibodies such as, but not limited to, bl2, 2F5, 2Gl 2, 4E10, M2909 either alone or combination, are mixed with purified HIV enables the antibodies to bind the glycoprotein spikes on the viral envelopes. The antibody-antigen complexes are dissociated, advantageously chemically dissociated, from the virus. The antibody-antigen complexes are purified and formulated into the vaccines of the present invention.

In yet another embodiment, new broadly neutralizing antibodies to HIV are identified and mixed with purified HIV enables the antibodies to bind the glycoprotein spikes on the viral envelopes. The antibody-antigen complexes are dissociated, advantageously chemically dissociated, from the virus. The antibody-antigen complexes are purified and formulated into the vaccines of the present invention.

In still another embodiment, the antibody-antigen complexes may be identified from alternate viral isolates, such as different HIV clades. In this embodiment, polyclonal anti- HIV sera, broadly neutralizing HIV monoclonal antibodies such as, but not limited to, bl2, 2F5, 2Gl 2, 4E10, M2909 either alone or combination, or newly identified broadly neutralizing antibodies to HIV are mixed with different HIV clade viral isolates to enable the antibodies to bind to varying antigens, thereby forming antibody-antigen complexes. The antibody-antigen complexes are dissociated, advantageously chemically dissociated, from the virus. The antibody-antigen complexes are purified and formulated into the vaccines of the present invention.

It is noted that in this disclosure and particularly in the claims and/or paragraphs, terms such as "comprises", "comprised", "comprising" and the like can have the meaning

attributed to it in U.S. Patent law; e.g., they can mean "includes", "included", "including", and the like; and that terms such as "consisting essentially of and "consists essentially of have the meaning ascribed to them in U.S. Patent law, e.g., they allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention.

These and other embodiments are disclosed or are obvious from and encompassed by, the following Detailed Description. DETAILED DESCRIPTION

The present invention relates to vaccines for HIV comprising antibody-antigen complexes. The current invention is based, in part, on Applicant's surprising discovery that immunization with antigen-antibody complexes elicit neutralizing antibody responses.

The present invention encompasses identification of antibody-antigen complexes for use as a HIV vaccine. In one embodiment, the invention relates to the identification of immunogenic antibody-antigen complexes. The invention encompasses mixing HIV antibodies, such as but not limited to, polyclonal anti-HIV sera, broadly neutralizing HIV monoclonal antibodies such as, but not limited to, bl2, 2F5, 2Gl 2, 4E10, M2909 either alone or combination or novel broadly neutralizing antibodies to HIV with purified HIV to enables the antibodies to bind to HIV antigens, such as but not limited to, the glycoprotein spikes on the viral envelopes, to form antibody-antigen complexes.

Any HIV antibody may be used for the binding to purified HIV to form antibody- antigen complexes. For example, the anti-HIV antibodies of U.S. Patent Nos. 6,949,337, 6,900,010, 6,821,744, 6,768,004, 6,613,743, 6,534,312, 6,511,830, 6,489,131, 6,242,197, 6,114,143, 6,074,646, 6,063,564, 6,060,254, 5,919,457, 5,916,806, 5,871,732, 5,824,304, 5,773,247, 5,736,320, 5,637,455, 5,587,285, 5,514,541, 5,317,009, 4,983,529, 4,886,742,

4,870,003 and 4,795,739 are useful for the present invention. Furthermore, monoclonal anti- HIV antibodies of U.S. Patent Nos. 7,074,556, 7,074,554, 7,070,787, 7,060,273, 7,045,130, 7,033,593, RE39,057, 7,008,622, 6,984,721, 6,972,126, 6,949,337, 6,946,465, 6,919,077, 6,916,475, 6,911,315, 6,905,680, 6,900,010, 6,825,217, 6,824,975, 6,818,392, 6,815,201, 6,812,026, 6,812,024, 6,797,811, 6,768,004, 6,703,019, 6,689,118, 6,657,050, 6,608,179, 6,600,023, 6,596,497, 6,589,748, 6,569,143, 6,548,275, 6,525,179, 6,524,582, 6,506,384, 6,498,006, 6,489,131, 6,465,173, 6,461,612, 6,458,933, 6,432,633, 6,410,318, 6,406,701, 6,395,275, 6,391,657, 6,391,635, 6,384,198, 6,376,170, 6,372,217, 6,344,545, 6,337,181, 6,329,202, 6,319,665, 6,319,500, 6,316,003, 6,312,931, 6,309,880, 6,296,807, 6,291,239,

6,261,558, 6,248,514, 6,245,331, 6,242,197, 6,241,986, 6,228,361, 6,221,580, 6,190,871, 6,177,253, 6,146,635, 6,146,627, 6,146,614, 6,143,876, 6,132,992, 6,124,132, RE36,866, 6,114,143, 6,103,238, 6,060,254, 6,039,684, 6,030,772, 6,020,468, 6,013,484, 6,008,044, 5,998,132, 5,994,515, 5,993,812, 5,985,545, 5,981,278, 5,958,765, 5,939,277, 5,928,930, 5,922,325, 5,919,457, 5,916,806, 5,914,109, 5,911,989, 5,906,936, 5,889,158, 5,876,716, 5,874,226, 5,872,012, 5,871,732, 5,866,694, 5,854,400, 5,849,583, 5,849,288, 5,840,480, 5,840,305, 5,834,599, 5,831,034, 5,827,723, 5,821,047, 5,817,767, 5,817,458, 5,804,440, 5,795,572, 5,783,670, 5,776,703, 5,773,225, 5,766,944, 5,753,503, 5,750,373, 5,747,641, 5,736,341, 5,731,189, 5,707,814, 5,702,707, 5,698,178, 5,695,927, 5,665,536, 5,658,745, 5,652,138, 5,645,836, 5,635,345, 5,618,922, 5,610,035, 5,607,847, 5,604,092, 5,601,819, 5,597,896, 5,597,688, 5,591,829, 5,558,865, 5,514,541, 5,510,264, 5,478,753, 5,374,518, 5,374,516, 5,344,755, 5,332,567, 5,300,433, 5,296,347, 5,286,852, 5,264,221, 5,260,308, 5,256,561, 5,254,457, 5,230,998, 5,227,159, 5,223,408, 5,217,895, 5,180,660, 5,173,399, 5,169,752, 5,166,050, 5,156,951, 5,140,105, 5,135,864, 5,120,640, 5,108,904, 5,104,790, 5,049,389, 5,030,718, 5,030,555, 5,004,697, 4,983,529, 4,888,290, 4,886,742 and 4,853,326, are also useful for the present invention.

In another embodiment, the antibody-antigen complexes may be identified from alternate viral isolates, such as different HIV clades (see, e.g., U.S. Provisional Patent Application No. 60/810,816, filed June 2, 2006, the disclosure of which is incorporated by reference). In this embodiment, polyclonal anti-HIV sera, broadly neutralizing HIV monoclonal antibodies such as, but not limited to, bl2, 2F5, 2Gl 2, 4E10, M2909 either alone or combination, or newly identified broadly neutralizing antibodies to HIV are mixed with different HIV clade viral isolates to enable the antibodies to bind to varying antigens, thereby forming antibody-antigen complexes. The antibody-antigen complexes are dissociated, advantageously chemically dissociated, preferably by solubilizing the HIV lipid bilayer, from the virus. In another embodiment, the antibody-antigen complexes may be dissociated with an affinity column, such as, but not limited to, CIq, Protein A or Protein G affinity columns or secondary antibodies. To purify antibody-antigen complexes, Protein A, Protein G, precipitating secondary antibodies or Protein A-bearing S. aureus cells may be used. The affinity of an antibody for Protein A or G is dependent on the subclass of the immunoglobulin and the species from which it comes. For example, Protein A is exceptionally well suited for immunoprecipitation of all rabbit primary antibodies, but not for chicken antibodies. To use Protein A for

immunoprecipitation of mouse primary antibodies, it is advisable to add 5 μg of rabbit anti- mouse IgG (secondary precipitating antibody) prior to the addition of Protein A/G (mix gently, and incubate for an additional 30 minutes at 4°C prior to adding Protein A/G). Following addition of Protein A/G agarose, incubate with gentle agitation for 30 minutes at 4°C, then wash at least three times by centrifugation and resuspension in immunoprecipitation buffer and collect antibody-antigen-Protein A/G complexes by centrifugation. The purified immune complex may be used for immunizations or other immunochemical techniques.

Antibody-antigen complexes as vaccine formulations are known in the art and the disclosures of any one of Akagaki & Inai (1983) MoI Immunol 20(11): 1221-6, Alber et al. (2000) Vaccine 19(7-8): 895-901, Andersson et al. (1995) Scand J Immunol 42(4): 407-17, Barr et al. (2003) Immunology 109(1): 87-92, Berger et al. (1996) Res Virol 147(2-3): 103-8, Berlyn et al. (2001) Clin Immunol 101(3): 276-83, Bonneau et al. (1972) Prog Immunobiol Stand 5: 537-41, Bouige et al. (1996) FEMS Immunol Med Microbiol 13(1): 71-9, Cannat et al. (1983) Ann Immunol (Paris) 134C(I): 43-53, Cavacini et al. (1995) J Immunol 155(7): 3638-44, Celis et al. (1987) Hepatology 7(3): 563-8, Chargelegue et al. (2005) Infect Immun 73(9): 5915-22, Dekker et al. (2004) MoI Biochem Parasitol 137(1): 143-9, Fenner (1972) Adv Exp Med Biol 31(0): 7-17, Genin & Lesavre (1983) MoI Immunol 20(10): 1069-72, Gnjatic et al. (2002) Proc Natl Acad Sci U S A 99(18): 11813-8, Guo et al. (2004) Avian Dis 48(1): 224-8, Habig et al. (1988) J Pediatr 112(1): 162-3, Haddad et al. (1997) Avian Dis

41(4): 882-9, Hanke et al. (1992) J Gen Virol 73 (Pt 3): 653-60, Hsueh et al. (1997) Cancer J Sci Am 3(6): 364-70, Ivan et al. (2001) Vet Immunol Immunopathol 79(3-4): 235-48, Ivan et al. (2005) Can J Vet Res 69(2): 135-42, Jeurissen et al. (1998) Immunology 95(3): 494-500, Kostiala & Kosunen (1972) Scand J Immunol 1(2): 143-51, Kraiselburd (1987) P R Health Sci J 6(1): 27-9, Kraiselburd et al. (1981) Infect Immun 33(2): 389-94, Kurul et al. (2004) Pediatr Nephrol 19(6): 621-6, Li et al. (2004) BMC Neurosci 5: 21, Lurhuma et al. (1994) East Afr Med J 71(8): 493-5, Marques et al. (2005) Clin Diagn Lab Immunol 12(9): 1036-40, McCluskie et al. (1998) Viral Immunol 11(4): 245-52, Montefiori et al. (1994) J Infect Dis 170(2): 429-32, Navol'nev (1983) Vestn Dermatol Venerol(l 1): 23-7, O'Lee et al. (1987) Arch Oral Biol 32(8): 539-43, Paccaud et al. (1987) Clin Exp Immunol 69(2): 468-76, Parish (1972) Immunology 22(6): 1087-98, Pizarro et al. (2002) Acta Crystallogr D Biol Crystallogr 58(Pt 7): 1246-8, Pokric et al. (1993) Vaccine 11(6): 655-9, Polack et al. (2002) J Exp Med 196(6): 859-65, Rafiq et al. (2002) J Clin Invest 110(1): 71-9, Randall et al. (1993) Vaccine 11(12): 1247-52, Randall et al. (1994) Vaccine 12(4): 351-8, Randall & Young (1988) J Gen

Virol 69 (Pt 10): 2505-16, Randall & Young (1989) J Virol 63(4): 1808-10, Randall et al. (1988) J Gen Virol 69 (Pt 10): 2517-26, Ridley et al. (1982) J Pathol 136(1): 59-72, Roic et al. (2006) J Vet Med B Infect Dis Vet Public Health 53(1): 17-23, Root-Bernstein (2004) J Clin Virol 31 Suppl 1 : S16-25, Schifferli et al. (1988) J Immunol 140(3): 899-904, Schnurr et al. (2005) Blood 105(6): 2465-72, Schultes & Nicodemus (2004) Expert Opin Biol Ther 4(8): 1265-84, Schuurhuis et al. (2006) J Immunol 176(8): 4573-80, Semkow & Wilczynski (1979) Acta Virol 23(1): 52-8, Speranskaia et al. (1998) Zh Mikrobiol Epidemiol Immunobiol(2): 14-8, Stager et al. (2003) Nat Med 9(10): 1287-92, Stoner et al. (1975) J Infect Dis 131(3): 230-8, Trkola et al. (1995) J Virol 69(11): 6609-17, van Rooijen (1975) Immunology 28(6): 1155-63, Wen et al. (1994) Chin Med J (Engl) 107(10): 741-4, Wen et al. (1999) Int Rev Immunol 18(3): 251-8, Wen et al. (1995) Lancet 345(8964): 1575-6, Whipple et al. (2006) MoI Immunol. 2006 Apr 20; [Epub ahead of print]

Wyss-Coray et al. (1992) Cell Immunol 139(1): 268-73, Yoshikawa et al. (2004) J Gen Virol 85(Pt 8): 2339-46 and Zheng et al. (2001) Vaccine 19(30): 4219-25 may be utilized for methods of the present invention.

The terms "protein", "peptide", "polypeptide", and "amino acid sequence" are used interchangeably herein to refer to polymers of amino acid residues of any length. The polymer may be linear or branched, it may comprise modified amino acids or amino acid analogs, and it may be interrupted by chemical moieties other than amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling or bioactive component.

As used herein, the terms "antigen" or "immunogen" are used interchangeably to refer to a substance, typically a protein, which is capable of inducing an immune response in a subject. The term also refers to proteins that are immunologically active in the sense that once administered to a subject (either directly or by administering to the subject a nucleotide sequence or vector that encodes the protein) is able to evoke an immune response of the humoral and/or cellular type directed against that protein. The term "antibody" includes intact molecules as well as fragments thereof, such as

Fab, F(ab')2, Fv and scFv which are capable of binding the epitopic determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and include, for example:

(i) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;

(ii) Fab', the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;

(iii) F(ab')2, the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab')2 is a dimer of two Fab' fragments held together by two disulfide bonds; (iv) scFv, including a genetically engineered fragment containing the variable region of a heavy and a light chain as a fused single chain molecule.

General methods of making these fragments are known in the art. (See for example, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York (1988), which is incorporated herein by reference). It should be understood that the proteins, including the antibodies and/or antigens of the invention may differ from the exact sequences illustrated and described herein. Thus, the invention contemplates deletions, additions and substitutions to the sequences shown, so long as the sequences function in accordance with the methods of the invention. In this regard, particularly preferred substitutions will generally be conservative in nature, i.e., those substitutions that take place within a family of amino acids. For example, amino acids are generally divided into four families: (1) acidic—aspartate and glutamate; (2) basic—lysine, arginine, histidine; (3) non-polar— alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar— glycine, asparagine, glutamine, cystine, serine threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified as aromatic amino acids. It is reasonably predictable that an isolated replacement of leucine with isoleucine or valine, or vice versa; an aspartate with a glutamate or vice versa; a threonine with a serine or vice versa; or a similar conservative replacement of an amino acid with a structurally related amino acid, will not have a major effect on the biological activity. Proteins having substantially the same amino acid sequence as the sequences illustrated and described but possessing minor amino acid substitutions that do not substantially affect the immunogenicity of the protein are, therefore, within the scope of the invention.

As used herein the terms "nucleotide sequences" and "nucleic acid sequences" refer to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) sequences, including, without limitation, messenger RNA (mRNA), DNA/RNA hybrids, or synthetic nucleic acids. The

nucleic acid can be single-stranded, or partially or completely double-stranded (duplex). Duplex nucleic acids can be homoduplex or heteroduplex.

As used herein the term "transgene" may used to refer to "recombinant" nucleotide sequences that may be derived from any of the nucleotide sequences encoding the proteins of the present invention. The term "recombinant" means a nucleotide sequence that has been manipulated "by man" and which does not occur in nature, or is linked to another nucleotide sequence or found in a different arrangement in nature. It is understood that manipulated "by man" means manipulated by some artificial means, including by use of machines, codon optimization, restriction enzymes, etc. For example, in one embodiment the nucleotide sequences may be mutated such that the activity of the encoded proteins in vivo is abrogated. In another embodiment the nucleotide sequences may be codon optimized, for example the codons may be optimized for human use. In preferred embodiments the nucleotide sequences of the invention are both mutated to abrogate the normal in vivo function of the encoded proteins, and codon optimized for human use. For example, each of the Gag, Pol, Env, Nef, RT, and Int sequences of the invention may be altered in these ways.

As regards codon optimization, the nucleic acid molecules of the invention have a nucleotide sequence that encodes the antigens of the invention and can be designed to employ codons that are used in the genes of the subject in which the antigen is to be produced. Many viruses, including HIV and other lentiviruses, use a large number of rare codons and, by altering these codons to correspond to codons commonly used in the desired subject, enhanced expression of the antigens can be achieved. In a preferred embodiment, the codons used are "humanized" codons, i.e., the codons are those that appear frequently in highly expressed human genes (Andre et al., J. Virol. 72:1497-1503, 1998) instead of those codons that are frequently used by HIV. Such codon usage provides for efficient expression of the transgenic HIV proteins in human cells. Any suitable method of codon optimization may be used. Such methods, and the selection of such methods, are well known to those of skill in the art. In addition, there are several companies that will optimize codons of sequences, such as Geneart (geneart.com). Thus, the nucleotide sequences of the invention can readily be codon optimized.

The invention further encompasses nucleotide sequences encoding functionally and/or antigenically equivalent variants and derivatives of the antigens of the invention and functionally equivalent fragments thereof. These functionally equivalent variants, derivatives, and fragments display the ability to retain antigenic activity. For instance,

changes in a DNA sequence that do not change the encoded amino acid sequence, as well as those that result in conservative substitutions of amino acid residues, one or a few amino acid deletions or additions, and substitution of amino acid residues by amino acid analogs are those which will not significantly affect properties of the encoded polypeptide. Conservative amino acid substitutions are glycine/alanine; valine/isoleucine/leucine; asparagine/glutamine; aspartic acid/glutamic acid; serine/threonine/methionine; lysine/arginine; and phenylalanine/tyrosine/tryptophan. In one embodiment, the variants have at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% homology or identity to the antigen, epitope, immunogen, peptide or polypeptide of interest.

For the purposes of the present invention, sequence identity or homology is determined by comparing the sequences when aligned so as to maximize overlap and identity while minimizing sequence gaps. In particular, sequence identity may be determined using any of a number of mathematical algorithms. A nonlimiting example of a mathematical algorithm used for comparison of two sequences is the algorithm of Karlin & Altschul, Proc. Natl. Acad. Sci. USA 1990; 87: 2264-2268, modified as in Karlin & Altschul, Proc. Natl. Acad. Sci. USA 1993;90: 5873-5877. Another example of a mathematical algorithm used for comparison of sequences is the algorithm of Myers & Miller, CABIOS 1988;4: 11-17. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM 120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Yet another useful algorithm for identifying regions of local sequence similarity and alignment is the FASTA algorithm as described in Pearson & Lipman, Proc. Natl. Acad. Sci. USA 1988; 85: 2444-2448.

Advantageous for use according to the present invention is the WU-BLAST (Washington University BLAST) version 2.0 software. WU-BLAST version 2.0 executable programs for several UNIX platforms can be downloaded from ftp

://blast.wustl.edu/blast/executables. This program is based on WU-BLAST version 1.4, which in turn is based on the public domain NCBI-BLAST version 1.4 (Altschul & Gish, 1996, Local alignment statistics, Doolittle ed., Methods in Enzymology 266: 460-480; Altschul et al, Journal of Molecular Biology 1990; 215: 403-410; Gish & States,

1993;Nature Genetics 3: 266-272; Karlin & Altschul, 1993;Proc. Natl. Acad. Sci. USA 90: 5873-5877; all of which are incorporated by reference herein).

The various recombinant nucleotide sequences and antibodies and/or antigens of the invention are made using standard recombinant DNA and cloning techniques. Such techniques are well known to those of skill in the art. See for example, "Molecular Cloning: A Laboratory Manual", second edition (Sambrook et al. 1989).

The nucleotide sequences of the present invention may be inserted into "vectors." The term "vector" is widely used and understood by those of skill in the art, and as used herein the term "vector" is used consistent with its meaning to those of skill in the art. For example, the term "vector" is commonly used by those skilled in the art to refer to a vehicle that allows or facilitates the transfer of nucleic acid molecules from one environment to another or that allows or facilitates the manipulation of a nucleic acid molecule.

Any vector that allows expression of the antibodies and/or antigens of the present invention may be used in accordance with the present invention. In certain embodiments, the antigens and/or antibodies of the present invention may be used in vitro (such as using cell- free expression systems) and/or in cultured cells grown in vitro in order to produce the encoded HIV- antigens and/or antibodies which may then be used for various applications such as in the production of proteinaceous vaccines. For such applications, any vector that allows expression of the antigens and/or antibodies in vitro and/or in cultured cells may be used.

For applications where it is desired that the antibodies and/or antigens be expressed in vivo, for example when the transgenes of the invention are used in DNA or DNA-containing vaccines, any vector that allows for the expression of the antibodies and/or antigens of the present invention and is safe for use in vivo may be used. In preferred embodiments the vectors used are safe for use in humans, mammals and/or laboratory animals.

For the antibodies and/or antigens of the present invention to be expressed, the protein coding sequence should be "operably linked" to regulatory or nucleic acid control sequences that direct transcription and translation of the protein. As used herein, a coding sequence and a nucleic acid control sequence or promoter are said to be "operably linked" when they are covalently linked in such a way as to place the expression or transcription and/or translation of the coding sequence under the influence or control of the nucleic acid control sequence. The "nucleic acid control sequence" can be any nucleic acid element, such as, but not limited to promoters, enhancers, IRES, introns, and other elements described herein that direct the expression of a nucleic acid sequence or coding sequence that is operably linked thereto. The

term "promoter" will be used herein to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II and that when operationally linked to the protein coding sequences of the invention lead to the expression of the encoded protein. The expression of the transgenes of the present invention can be under the control of a constitutive promoter or of an inducible promoter, which initiates transcription only when exposed to some particular external stimulus, such as, without limitation, antibiotics such as tetracycline, hormones such as ecdysone, or heavy metals. The promoter can also be specific to a particular cell-type, tissue or organ. Many suitable promoters and enhancers are known in the art, and any such suitable promoter or enhancer may be used for expression of the transgenes of the invention. For example, suitable promoters and/or enhancers can be selected from the Eukaryotic Promoter Database (EPDB).

The vectors used in accordance with the present invention should typically be chosen such that they contain a suitable gene regulatory region, such as a promoter or enhancer, such that the antigens and/or antibodies of the invention can be expressed. For example, when the aim is to express the antibodies and/or antigens of the invention in vitro, or in cultured cells, or in any prokaryotic or eukaryotic system for the purpose of producing the protein(s) encoded by that antibody and/or antigen, then any suitable vector can be used depending on the application. For example, plasmids, viral vectors, bacterial vectors, protozoal vectors, insect vectors, baculovirus expression vectors, yeast vectors, mammalian cell vectors, and the like, can be used. Suitable vectors can be selected by the skilled artisan taking into consideration the characteristics of the vector and the requirements for expressing the antibodies and/or antigens under the identified circumstances.

When the aim is to express the antibodies and/or antigens of the invention in vivo in a subject, for example in order to generate an immune response against an HIV-I antigen and/or protective immunity against HIV-I, expression vectors that are suitable for expression on that subject, and that are safe for use in vivo, should be chosen. For example, in some embodiments it may be desired to express the antibodies and/or antigens of the invention in a laboratory animal, such as for pre-clinical testing of the HIV-I immunogenic compositions and vaccines of the invention. In other embodiments, it will be desirable to express the antibodies and/or antigens of the invention in human subjects, such as in clinical trials and for actual clinical use of the immunogenic compositions and vaccine of the invention. Any vectors that are suitable for such uses can be employed, and it is well within the capabilities of the skilled artisan to selelct a suitable vector. In some embodiments it may be preferred

that the vectors used for these in vivo applications are attenuated to vector from amplifying in the subject. For example, if plasmid vectors are used, preferably they will lack an origin of replication that functions in the subject so as to enhance safety for in vivo use in the subject.. If viral vectors are used, preferably they are attenuated or replication-defective in the subject, again, so as to enhance safety for in vivo use in the subject.

In preferred embodiments of the present invention viral vectors are used. Viral expression vectors are well known to those skilled in the art and include, for example, viruses such as adenoviruses, adeno-associated viruses (AAV), alphaviruses, herpesviruses, retroviruses and poxviruses, including avipox viruses, attenuated poxviruses, vaccinia viruses, and particularly, the modified vaccinia Ankara virus (MVA; ATCC Accession No. VR- 1566). Such viruses, when used as expression vectors are innately non-pathogenic in the selected subjects such as humans or have been modified to render them non-pathogenic in the selected subjects. For example, replication-defective adenoviruses and alphaviruses are well known and can be used as gene delivery vectors. In particularly preferred embodiments adenovirus vectors are used. Many adenovirus vectors are known in the art and any such suitable vector my be used. In preferred embodiments the adenovirus vector used is selected from the group consisting of the Ad5, Ad35, AdI 1, C6, and C7 vectors.

The sequence of the Adenovirus 5 ("Ad5") genome has been published. (Chroboczek, J., Bieber, F., and Jacrot, B. (1992) The Sequence of the Genome of Adenovirus Type 5 and Its Comparison with the Genome of Adenovirus Type 2, Virology 186, 280-285; the contents if which is hereby incorporated by reference). Ad35 vectors are described in U.S. Patent Nos. 6,974,695, 6,913,922, and 6,869,794. AdI 1 vectors are described in U.S. Patent No. 6,913,922. C6 adenovirus vectors are described in U.S. Patent Nos. 6,780,407; 6,537,594; 6,309,647; 6,265,189; 6,156,567; 6,090,393; 5,942,235 and 5,833,975. C7 vectors are described in U.S. Patent No. 6,277,558.

Adenovirus vectors that are El -defective or deleted, E3 -defective or deleted, and/or E4-defective or deleted may also be used. Certain adenoviruses having mutations in the El region have improved safety margin because El -defective adenovirus mutants are replication-defective in non-permissive cells, or, at the very least, are highly attenuated.

Adenoviruses having mutations in the E3 region may have enhanced the immunogenicity by disrupting the mechanism whereby adenovirus down-regulates MHC class I molecules. Adenoviruses having E4 mutations may have reduced immunogenicity of the adenovirus vector because of suppression of late gene expression. Such vectors may be particularly

useful when repeated re-vaccination utilizing the same vector is desired. Adenovirus vectors that are deleted or mutated in El, E3, E4, El and E3, and El and E4 can be used in accordance with the present invention.

Furthermore, "gutless" adenovirus vectors, in which all viral genes are deleted, can also be used in accordance with the present invention. Such vectors require a helper virus for their replication and require a special human 293 cell line expressing both EIa and Cre, a condition that does not exist in natural environment. Such "gutless" vectors are non- immunogenic and thus the vectors may be inoculated multiple times for re-vaccination. The "gutless" adenovirus vectors can be used for insertion of heterologous inserts/genes such as the transgenes of the present invention, and can even be used for co-delivery of a large number of heterologous inserts/genes.

The nucleotide sequences and vectors of the invention can be delivered to cells, for example if aim is to express and the HIV-I antigens in cells in order to produce and isolate the expressed proteins, such as from cells grown in culture. For expressing the antibodies and/or antigens in cells any suitable transfection, transformation, or gene delivery methods can be used. Such methods are well known by those skilled in the art, and one of skill in the art would readily be able to select a suitable method depending on the nature of the nucleotide sequences, vectors, and cell types used. For example, transfection, transformation, microinjection, infection, electroporation, lipofection, or liposome-mediated delivery could be used. Expression of the antibodies and/or antigens can be carried out in any suitable type of host cells, such as bacterial cells, yeast, insect cells, and mammalian cells. The antibodies and/or antigens of the invention can also be expressed using including in vitro transcription/translation systems. All of such methods are well known by those skilled in the art, and one of skill in the art would readily be able to select a suitable method depending on the nature of the nucleotide sequences, vectors, and cell types used.

Following expression, the antibodies and/or antigens of the invention can be isolated and/or purified or concentrated using any suitable technique known in the art. For example, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, immuno-affϊnity chromatography, hydroxyapatite chromatography, lectin chromatography, molecular sieve chromatography, isoelectric focusing, gel electrophoresis, or any other suitable method or combination of methods can be used.

In preferred embodiments, the nucleotide sequences, antibodies and/or antigens of the invention are administered in vivo, for example where the aim is to produce an immunogenic

response in a subject. A "subject" in the context of the present invention may be any animal. For example, in some embodiments it may be desired to express the transgenes of the invention in a laboratory animal, such as for pre-clinical testing of the HIV-I immunogenic compositions and vaccines of the invention. In other embodiments, it will be desirable to express the antibodies and/or antigens of the invention in human subjects, such as in clinical trials and for actual clinical use of the immunogenic compositions and vaccine of the invention. In preferred embodiments the subject is a human, for example a human that is infected with, or is at risk of infection with, HIV-I .

For such in vivo applications the nucleotide sequences, antibodies and/or antigens of the invention_are preferably administered as a component of an immunogenic composition comprising the nucleotide sequences and/or antigens of the invention in admixture with a pharmaceutically acceptable carrier. The immunogenic compositions of the invention are useful to stimulate an immune response against HIV-I and may be used as one or more components of a prophylactic or therapeutic vaccine against HIV-I for the prevention, amelioration or treatment of AIDS. The nucleic acids and vectors of the invention are particularly useful for providing genetic vaccines, i.e. vaccines for delivering the nucleic acids encoding the antibodies and/or antigens of the invention to a subject, such as a human, such that the antibodies and/or antigens are then expressed in the subject to elicit an immune response. The compositions of the invention may be injectable suspensions, solutions, sprays, lyophilized powders, syrups, elixirs and the like. Any suitable form of composition may be used. To prepare such a composition, a nucleic acid or vector of the invention, having the desired degree of purity, is mixed with one or more pharmaceutically acceptable carriers and/or excipients. The carriers and excipients must be "acceptable" in the sense of being compatible with the other ingredients of the composition. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, or combinations thereof, buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino

acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG).

An immunogenic or immunological composition can also be formulated in the form of an oil-in- water emulsion. The oil-in- water emulsion can be based, for example, on light liquid paraffin oil (European Pharmacopea type); isoprenoid oil such as squalane, squalene, EICOSANE ™ or tetratetracontane; oil resulting from the oligomerization of alkene(s), e.g., isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, such as plant oils, ethyl oleate, propylene glycol di(caprylate/caprate), glyceryl tri(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, e.g., isostearic acid esters. The oil advantageously is used in combination with emulsifϊers to form the emulsion. The emulsifϊers can be nonionic surfactants, such as esters of sorbitan, mannide (e.g., anhydromannitol oleate), glycerol, polyglycerol, propylene glycol, and oleic, isostearic, ricinoleic, or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene- polyoxyethylene copolymer blocks, such as the Pluronic® products, e.g., L121. The adjuvant can be a mixture of emulsifϊer(s), micelle-forming agent, and oil such as that which is commercially available under the name Provax® (IDEC Pharmaceuticals, San Diego, CA). The immunogenic compositions of the invention can contain additional substances, such as wetting or emulsifying agents, buffering agents, or adjuvants to enhance the effectiveness of the vaccines (Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Company, (ed.) 1980).

Adjuvants may also be included. Adjuvants include, but are not limited to, mineral salts (e.g., A1K(SO 4 ) 2 , AlNa(SO 4 ) 2 , A1NH(SO 4 ) 2 , silica, alum, Al(OH) 3 , Ca 3 (PO 4 ) 2 , kaolin, or carbon), polynucleotides with or without immune stimulating complexes (ISCOMs) (e.g., CpG oligonucleotides, such as those described in Chuang, T.H. et al, (2002) J. Leuk. Biol. 71(3): 538-44; Ahmad-Nejad, P. et al (2002) Eur. J. Immunol. 32(7): 1958-68; poly IC or poly AU acids, polyarginine with or without CpG (also known in the art as IC31; see Schellack, C. et al (2003) Proceedings of the 34 th Annual Meeting of the German Society of Immunology; Lingnau, K. et al (2002) Vaccine 20(29-30): 3498-508), JuvaVax™ (U.S. Patent No. 6,693,086), certain natural substances (e.g., wax D from Mycobacterium tuberculosis, substances found in Corny ebacterium parvum, Bordetella pertussis, or members

of the genus Brucella), flagellin (Toll-like receptor 5 ligand; see McSorley, S.J. et al (2002) J. Immunol. 169(7): 3914-9), saponins such as QS21, QS17, and QS7 (U.S. Patent Nos. 5,057,540; 5,650,398; 6,524,584; 6,645,495), monophosphoryl lipid A, in particular, 3-de-O- acylated monophosphoryl lipid A (3D-MPL), imiquimod (also known in the art as IQM and commercially available as Aldara®; U.S. Patent Nos. 4,689,338; 5,238,944; Zuber, A.K. et al (2004) 22(13-14): 1791-8), and the CCR5 inhibitor CMPD167 (see Veazey, R.S. et al (2003) J. Exp. Med. 198: 1551-1562).

Aluminum hydroxide or phosphate (alum) are commonly used at 0.05 to 0.1% solution in phosphate buffered saline. Other adjuvants that can be used, especially with DNA vaccines, are cholera toxin, especially CTAl-DD/ISCOMs (see Mowat, A.M. et al (2001) J. Immunol. 167(6): 3398-405), polyphosphazenes (Allcock, H.R. (1998) App. Organometallic Chem. 12(10-11): 659-666; Payne, L.G. et al (1995) Pharm. Biotechnol. 6: 473-93), cytokines such as, but not limited to, IL-2, IL-4, GM-CSF, IL-12, IL-15 IGF-I, IFN-α, IFN- β, and IFN-γ (Boyer et al., (2002) J. Liposome Res. 121 :137-142; WO01/095919), immunoregulatory proteins such as CD40L (ADX40; see, for example, WO03/063899), and the CDIa ligand of natural killer cells (also known as CRONY or α-galactosyl ceramide; see Green, T.D. et al, (2003) J. Virol. 77(3): 2046-2055), immunostimulatory fusion proteins such as IL-2 fused to the Fc fragment of immunoglobulins (Barouch et al., Science 290:486- 492, 2000) and co-stimulatory molecules B7.1 and B7.2 (Boyer), all of which can be administered either as proteins or in the form of DNA, on the same expression vectors as those encoding the antigens of the invention or on separate expression vectors.

The immunogenic compositions can be designed to introduce the antibodies, antigens, antibody-antigen complexes, nucleic acids or expression vectors to a desired site of action and release it at an appropriate and controllable rate. Methods of preparing contra lled-release formulations are known in the art. For example, controlled release preparations can be produced by the use of polymers to complex or absorb the immunogen and/or immunogenic composition. A controlled-release formulations can be prepared using appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) known to provide the desired controlled release characteristics or release profile. Another possible method to control the duration of action by a controlled-release preparation is to incorporate the active ingredients into particles of a polymeric material such as, for example, polyesters, polyamino acids, hydrogels, polylactic acid, polyglycolic acid, copolymers of these acids, or

ethylene vinylacetate copolymers. Alternatively, instead of incorporating these active ingredients into polymeric particles, it is possible to entrap these materials into microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacrylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in New Trends and Developments in Vaccines, Voller et al. (eds.), University Park Press, Baltimore, Md., 1978 and Remington's Pharmaceutical Sciences, 16th edition. Suitable dosages of the antibodies, antigens, antibody-antigen complexes, nucleic acids and expression vectors of the invention (collectively, the immunogens) in the immunogenic composition of the invention can be readily determined by those of skill in the art. For example, the dosage of the immunogens can vary depending on the route of administration and the size of the subject. Suitable doses can be determined by those of skill in the art, for example by measuring the immune response of a subject, such as a laboratry animal, using conventional immunological techniques, and adjusting the dosages as appropriate. Such techniques for measuring the immune response of the subject include but are not limited to, chromium release assays, tetramer binding assays, IFN-γ ELISPOT assays, IL-2 ELISPOT assays, intracellular cytokine assays, and other immunological detection assays, e.g., as detailed in the text "Antibodies: A Laboratory Manual" by Ed Harlow and David Lane.

When provided prophylactically, the immunogenic compositions of the invention are ideally administered to a subject in advance of HIV infection, or evidence of HIV infection, or in advance of any symptom due to AIDS, especially in high-risk subjects. The prophylactic administration of the immunogenic compositions can serve to provide protective immunity of a subject against HIV-I infection or to prevent or attenuate the progression of AIDS in a subject already infected with HIV-I . When provided therapeutically, the immunogenic compositions can serve to ameliorate and treat AIDS symptoms and are advantageously used as soon after infection as possible, preferably before appearance of any symptoms of AIDS but may also be used at (or after) the onset of the disease symptoms. The immunogenic compositions can be administered using any suitable delivery method including, but not limited to, intramuscular, intravenous, intradermal, mucosal, and topical delivery. Such techniques are well known to those of skill in the art. More specific

examples of delivery methods are intramuscular injection, intradermal injection, and subcutaneous injection. However, delivery need not be limited to injection methods. Further, delivery of DNA to animal tissue has been achieved by cationic liposomes (Watanabe et al, (1994) MoI. Reprod. Dev. 38:268-274; and WO 96/20013), direct injection of naked DNA into animal muscle tissue (Robinson et al., (1993) Vaccine 11 :957-960;

Hoffman et al., (1994) Vaccine 12: 1529-1533; Xiang et al., (1994) Virology 199: 132-140; Webster et al., (1994) Vaccine 12: 1495-1498; Davis et al., (1994) Vaccine 12: 1503-1509; and Davis et al., (1993) Hum. MoI. Gen. 2: 1847-1851), or intradermal injection of DNA using "gene gun" technology (Johnston et al., (1994) Meth. Cell Biol. 43:353-365). Alternatively, delivery routes can be oral, intranasal or by any other suitable route. Delivery also be accomplished via a mucosal surface such as the anal, vaginal or oral mucosa. Immunization schedules (or regimens) are well known for animals (including humans) and can be readily determined for the particular subject and immunogenic composition. Hence, the immunogens can be administered one or more times to the subject. Preferably, there is a set time interval between separate administrations of the immunogenic composition. While this interval varies for every subject, typically it ranges from 10 days to several weeks, and is often 2, 4, 6 or 8 weeks. For humans, the interval is typically from 2 to 6 weeks. The immunization regimes typically have from 1 to 6 administrations of the immunogenic composition, but may have as few as one or two or four. The methods of inducing an immune response can also include administration of an adjuvant with the immunogens. In some instances, annual, biannual or other long interval (5-10 years) booster immunization can supplement the initial immunization protocol.

The present methods also include a variety of prime-boost regimens, especially DNA prime- Adenovirus boost regimens. In these methods, one or more priming immunizations are followed by one or more boosting immunizations. The actual immunogenic composition can be the same or different for each immunization and the type of immunogenic composition (e.g., containing protein or expression vector), the route, and formulation of the immunogens can also be varied. For example, if an expression vector is used for the priming and boosting steps, it can either be of the same or different type (e.g., DNA or bacterial or viral expression vector). One useful prime-boost regimen provides for two priming immunizations, four weeks apart, followed by two boosting immunizations at 4 and 8 weeks after the last priming immunization. It should also be readily apparent to one of skill in the art that there are several permutations and combinations that are encompassed using the DNA, bacterial and viral expression vectors of the invention to provide priming and boosting regimens.

A specific embodiment of the invention provides methods of inducing an immune response against HIV in a subject by administering an immunogenic composition of the invention, preferably comprising an adenovirus vector containing DNA encoding one or more of the antibodies, antigens, antibody-antigen complexes of the invention, one or more times to a subject wherein the antibodies, antigens, antibody-antigen complexes are expressed at a level sufficient to induce a specific immune response in the subject. Such immunizations can be repeated multiple times at time intervals of at least 2, 4 or 6 weeks (or more) in accordance with a desired immunization regime.

The immunogenic compositions of the invention can be administered alone, or can be co-administered, or sequentially administered, with other HIV immunogens and/or HIV immunogenic compositions, e.g., with "other" immunological, antigenic or vaccine or therapeutic compositions thereby providing multivalent or "cocktail" or combination compositions of the invention and methods of employing them. Again, the ingredients and manner (sequential or co-administration) of administration, as well as dosages can be determined taking into consideration such factors as the age, sex, weight, species and condition of the particular subject, and the route of administration.

When used in combination, the other HIV immunogens can be administered at the same time or at different times as part of an overall immunization regime, e.g., as part of a prime-boost regimen or other immunization protocol. In an advantageous embodiment, the other HIV immunogen is env, preferably the HIV env trimer.

Many other HIV immunogens are known in the art, one such preferred immunogen is HIVA (described in WO 01/47955), which can be administered as a protein, on a plasmid (e.g., pTHr.HIVA) or in a viral vector (e.g., MVA.HIVA). Another such HIV immunogen is RENTA (described in PCT/US2004/037699), which can also be administered as a protein, on a plasmid (e.g., pTHr.RENTA) or in a viral vector (e.g., MVA.RENTA).

For example, one method of inducing an immune response against HIV in a human subject comprises administering at least one priming dose of an HIV immunogen and at least one boosting dose of an HIV immunogen, wherein the immunogen in each dose can be the same or different, provided that at least one of the immunogens is an antibody, antigen or antibody-antigen complex of the present invention, a nucleic acid encoding an antibody, antigen or antibody-antigen complex of the invention or an expression vector, preferably an adenovirus vector, encoding an antibody, antigen or antibody-antigen complex of the invention, and wherein the immunogens are administered in an amount or expressed at a level sufficient to induce an HIV-specific immune response in the subject. The HIV-specific

immune response can include an HIV-specifϊc T-cell immune response or an HIV-specific B- cell immune response. Such immunizations can be done at intervals, preferably of at least 2- 6 or more weeks.

It is to be understood and expected that variations in the principles of invention as described above may be made by one skilled in the art and it is intended that such modifications, changes, and substitutions are to be included within the scope of the present invention.

The invention is further described by the following numbered paragraphs:

1. A method of producing an immune response comprising administering to a mammal a purified antibody-antigen complex dissociated from polyclonal anti-HIV sera bound to glycoprotein spikes on HIV envelopes.

2. A method of producing an immune response comprising administering to a mammal a purified antibody-antigen complex dissociated from a mixture of broadly neutralizing antibodies and HIV, wherein the mixture is bound to glycoprotein spikes on HIV envelopes.

3. The method of paragraph 2 wherein the antibodies are monoclonal antibodies.

4. The method of paragraph 3 wherein the monoclonal antibodies are bl2, 2F5, 2Gl 2, 4E10, M2909 or any combination thereof.

5. The method of any one of paragraphs 2-4 wherein the HIV is purified HIV. 6. The method of any one of paragraphs 2-4 wherein the HIV is a HIV viral isolate.

7. The method of paragraph 6 wherein the HIV viral isolate is a HIV clade viral isolate.

8. The method of any one of paragraphs 1-7 wherein the purified antibody- antigen complex is chemically dissociated from the glycoprotein spikes.

9. The method of any one of paragraphs 1-8 wherein the purified antibody- antigen complex is dissociated from the glycoprotein spikes by solubilizing a HIV lipid bilayer.

10. The method of any one of paragraphs 1-7 wherein the purified antibody- antigen complex is purified with Protein A, protein G, precipitating secondary antibodies or Protein A-bearing S. aureus cells.

11. The method of any one of paragraphs 1-10 wherein the mammal is a human.

12. The method of any one of paragraphs 1-11 wherein the purified antibody- antigen complex is administered in a pharmaceutically acceptable carrier.

13. The method of any one of paragraphs 1-12 wherein the administering further comprises a prime-boost regimen.

Having thus described in detail preferred embodiments of the present invention, it is to be understood that the invention defined by the above paragraphs is not to be limited to particular details set forth in the above description as many apparent variations thereof are possible without departing from the spirit or scope of the present invention.