Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
USING HEAT SHOCK PROTEINS TO INCREASE IMMUNE RESPONSE
Document Type and Number:
WIPO Patent Application WO/2003/090687
Kind Code:
A2
Abstract:
The present invention provides for a method of using heat shock proteins (HSPs) or alpha-2-macroglobulins (&agr 2Ms) to amplify the immune response initiated by a vaccine. HSP or &agr 2M can be introduced into a subject before, concurrently, or after the administration of a vaccine. The HSP or &agr 2M can also be used to activate antigen presenting cells which are then introduced into a subject in conjunction with a vaccine. The HSP or &agr 2M used in the methods of the invention can be unbound or can be covalently or noncovalently bound to a peptide that is unrelated to the vaccine. The subject is preferably mammalian, and most preferably human. It is shown by way of example herein that HSPs induces secretion of cytokines and surface expression of antigen-presenting and co­stimulatory molecules. The invention also encompasses methods of treatment and prevention of cancer and infectious diseases in a subject.

Inventors:
SRIVASTAVA PRAMOD K
Application Number:
PCT/US2003/012803
Publication Date:
November 06, 2003
Filing Date:
April 25, 2003
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CONNECTICUT HEALTH CT (US)
International Classes:
A61K45/00; A61K38/17; A61K39/00; A61K39/005; A61K39/008; A61K39/012; A61K39/02; A61K39/04; A61K39/095; A61K39/118; A61K39/12; A61K39/13; A61K39/145; A61K39/165; A61K39/20; A61K39/21; A61K39/235; A61K39/245; A61K39/25; A61K39/29; A61K39/39; A61P31/04; A61P35/00; A61P35/02; A61P43/00; C07K14/47; (IPC1-7): A61K/
Foreign References:
US6017540A2000-01-25
Other References:
CHU C.T. ET AL: 'Adjuvant-free in vivo targeting. Antigen delivery by alpha2-macroglobulin enhances antibody formation' JOURNAL OF IMMONOLOGY vol. 152, no. 4, February 1994, pages 1538 - 1545, XP002123497
CIANCIOLO G.J. ET AL: 'Covalent complexes of antigen and alpha2-macroglobulin: evidence for dramatically-increased immunogenicity' VACCINE vol. 20, no. 3-4, November 2001, pages 554 - 562, XP004310165
ZHANG W.M. ET AL: 'Characterization And Immunological Determination Of The Complex Between Prostate-Specific Antigen And A2-Macroglobulin' CLINICAL CHEMISTRY vol. 44, no. 12, December 1998, pages 2471 - 2479, XP002919882
Attorney, Agent or Firm:
Antler, Adriane M. (222 East 41st Street New York, NY, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. A method for producing an immune response in a subject comprising the steps of : (a) administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced; and (b) administering to the subject an o2M preparation, wherein the o2M preparation does not display the immunogenicity of the component, such that an immune response to the component is produced in the subject.
2. A method of inducing an immune response by a vaccine composition in a subject comprising the steps of : (a) administering to the subject an o2M preparation; and (b) administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced, the vaccine composition being in an amount that is sub immunogenic for the component in the absence of step (a), such that an immune response to the component is induced in the subject, and wherein the o2M preparation does not display the immunogenicity of the component.
3. A method of treating or preventing an infectious disease in a subject comprising the steps of : (a) administering to the subject a vaccine composition comprising a component that displays the antigenicity of an infectious agent that causes the infectious disease; and (b) administering to the subject an amount of an o2M preparation effective in combination with step (a) to induce or increase an immune response to the component in the subject, wherein the o2M preparation does not display the immunogenicity of the component.
4. A method of treating or preventing a cancer in a subject comprising the steps of : (a) administering to the subject a vaccine composition comprising a component that displays the antigenicity of a cancer cell; and (b) administering to the subject an amount of an o2M preparation effective to induce or increase an immune response in the subject to the component, wherein the c2M preparation does not display the immunogenicity of the component.
5. The method of claim 1, wherein the immune response to the component produced in the subject is increased relative to the immune response to the component in the subject in the absence of step (b).
6. The method according to claim 1 wherein the o2M preparation comprises o2Mpeptide complexes.
7. The method according to claim 2 wherein the o2M preparation comprises a2Mpeptide complexes.
8. The method according to claim 3 wherein the o2M preparation comprises o2Mpeptide complexes.
9. The method according to claim 4 wherein the o2M preparation comprises o2Mpeptide complexes.
10. The method according to claim 1 wherein the o2M preparation comprises purified oe2M.
11. The method according to claim 2 wherein the o2M preparation comprises purified o2M.
12. The method according to claim 3 wherein the o2M preparation comprises purified o2M.
13. The method according to claim 4 wherein the odM preparation comprises purified o2M.
14. The method according to claim 1 wherein the o2M preparation comprises o2Mpeptide complexes and purified ou2M.
15. The method according to claim 2 wherein the a2M preparation comprises o2Mpeptide complexes and purified oe2M.
16. The method according to claim 3 wherein the o2M preparation comprises o2Mpeptide complexes and purified o2M.
17. The method according to claim 4 wherein the o2M preparation comprises o2Mpeptide complexes and purified o2M.
18. The method according to claim 1 wherein the subject is human and the o2M preparation comprises mammalian oe2M.
19. The method according to claim 2 wherein the subject is human and the Q2M preparation comprises mammalian o2M.
20. The method according to claim 3 wherein the subject is human and the o2M preparation comprises mammalian o2M.
21. The method according to claim 4 wherein the subject is human and the o2M preparation comprises mammalian o2M.
22. The method according to claim 1,2, 3, or 4 wherein the o2M preparation is administered before the administration of the vaccine composition.
23. The method according to claim 1,2, 3, or 4 wherein the o2M preparation is administered concurrently with the administration of the vaccine composition.
24. The method according to claim 1,2, 3, or 4 wherein the o2M preparation is administered after the administration of the vaccine composition.
25. The method according to claim 6,7, 8, or 9 wherein the o2M preparation is administered before the administration of the vaccine composition.
26. The method according to claim 6,7, 8, or 9 wherein the o2M preparation is administered concurrently with the administration of the vaccine composition.
27. The method according to claim 6,7, 8, or 9 wherein the o2M preparation is administered after the administration of the vaccine composition.
28. The method according to claim 10,11, 12, or 13 wherein the o2M preparation is administered before the administration of the vaccine composition.
29. The method according to claim 10,11, 12, or 13 wherein the o2M preparation is administered concurrently with the administration of the vaccine composition.
30. The method according to claim 10,11, 12, or 13 wherein the o2M preparation is administered after the administration of the vaccine composition.
31. The method according to claim 14,15, 16, or 17 wherein the o2M preparation is administered before the administration of the vaccine composition.
32. The method according to claim 14,15, 16, or 17 wherein the o2M preparation is administered concurrently with the administration of the vaccine composition.
33. The method according to claim 14,15, 16, or 17 wherein the o2M preparation is administered after the administration of the vaccine composition.
34. The method according to claim 14, 15, 16, or 17 wherein the o2M preparation and the vaccine composition are both administered on the same day.
35. The method of claim 18, 19,20, or 21 wherein the vaccine composition is a live vaccine, an attenuated vaccine, a subunit vaccine, a DNA vaccine, or a RNA vaccine.
36. The method according to claim 3 wherein the infectious disease is selected from the group consisting of hepatitis A virus, hepatitis B virus, hepatitis C virus, influenza, varicella, adenovirus, herpes simplex I virus, herpes simplex II virus, rinderpest, rhinovirus, ECHO virus, rotavirus, respiratory syncytial virus, papilloma virus, papova virus, cytomegalovirus, echinovirus, arbovirus, hantavirus, coxsackie virus, mumps virus, measles virus, rubella virus, polio virus, human immunodeficiency virus type I (HIVI), human immunodeficiency virus type II (HIVII), mycobacteria, rickettsia, mycoplasma, neisseria, legionella, leishmania, kokzidioa, trypanosoma and chlamydia.
37. The method according to claim 4 wherein the cancer is selected from the group consisting of fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms'tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma; leukemias, e. g., acute lymphocytic leukemia and acute myelocytic leukemia (myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); and polycythemia vera, lymphom (Hodgkin's disease and nonHodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, and heavy chain disease.
38. The method of claim 3, wherein the method is for preventing an infectious disease.
39. The method of claim 4, wherein the method is for treating a cancer.
40. A kit comprising: (a) a first container containing an o2M preparation in an amount effective to increase an immune response elicited by a vaccine composition against a component of the vaccine composition against which an immune response is desired; and (b) a second container containing the vaccine composition in an amount that, when administered before, concurrently with, or after the administration of the cm preparation of (a), is effective to induce an immune response against the component.
41. The kit according to claim 40 wherein the o2M preparation comprises o2Mpeptide complexes.
42. A method for improving the outcome of a treatment using a therapeutic modality in a subject comprising administering to a subject receiving the therapeutic modality a mammalian c2M preparation, wherein the therapeutic modality is not a vaccine.
43. The method of claim 42 wherein the treatment modality is an antibiotic, an antiviral agent, an antifungal agent, a chemotherapeutic agent, or radiation.
44. The method of claim 43 wherein the subject is human and the o2M preparation comprises human o2M.
45. The method of claim 1,2, 3, or 4, wherein said vaccine composition does not comprise a heat shock protein or an o2M.
46. The method of claim 1,2, 3, or 4, with the proviso that when said component of the vaccine composition is a peptide complexed to a heat shock protein or to oe2M, said vaccine composition and said o2M preparation are not present in admixture.
47. A method for producing an immune response in a subject comprising the steps of : (a) administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced; and (b) administering to the subject a heat shock protein preparation, wherein the heat shock protein preparation does not display the immunogenicity of the component, such that an immune response to the component is produced in the subject, with the proviso that when said component of the vaccine composition is a peptide complexed to a heat shock protein or an o2M, said vaccine composition and said heat shock protein preparation are not present in admixture.
48. A method of inducing an immune response by a vaccine composition in a subject comprising the steps of : (a) administering to the subject a heat shock protein preparation, wherein the heat shock protein preparation does not display the immunogenicity of a component against which an immune response is desired to be induced; and (b) administering to the subject a vaccine composition comprising the component against which an immune response is desired to be induced, the vaccine composition being in an amount that is sub immunogenic for the component in the absence of step (a), such that an immune response to the component is induced in the subject, with the proviso that when said component of the vaccine composition is a peptide complexed to a heat shock protein or an a2M, said vaccine composition and said heat shock protein preparation are not present in admixture.
49. A method of treating or preventing an infectious disease in a subject comprising the steps of : (a) administering to the subject a vaccine composition comprising a component that displays the antigenicity of an antigen of an infectious agent that causes the infectious disease; and (b) administering to the subject an amount of a heat shock protein preparation effective in combination with step (a) to induce or increase an immune response to the component in the subject, wherein the heat shock protein preparation does not display the immunogenicity of the component, with the proviso that when said component of the vaccine composition is a peptide complexed to a heat shock protein or an c2M, said vaccine composition and said heat shock protein preparation are not present in admixture.
50. A method of treating or preventing a cancer in a subject comprising the steps of : (a) administering to the subject a vaccine composition comprising a component that displays the antigenicity of a tumor specific or tumor associated antigen of a cancer cell; and (b) administering to the subject an amount of a heat shock protein preparation effective to induce or increase an immune response in the subject to the component, wherein the heat shock protein preparation does not display the immunogenicity of the component, with the proviso that when said component of the vaccine composition is a peptide complexed to a heat shock protein or an o2M, said vaccine composition and said heat shock protein preparation are not present in admixture.
51. The method of claim 47 wherein said component of the vaccine composition is a peptide complexed to a heat shock protein or an o2M.
52. The method of claim 48 wherein said component of the vaccine composition is a peptide complexed to a heat shock protein or an o2M.
53. The method of claim 49 wherein said component of the vaccine composition is a peptide complexed to a heat shock protein or an o2M.
54. The method of claim 50 wherein said component of the vaccine composition is a peptide complexed to a heat shock protein or an o2M.
Description:
USING HEAT SHOCK PROTEINS TO INCREASE IMMUNE RESPONSE This application is a continuation-in-part of co-pending U. S. Application Serial No. 09/693,643, filed October 20,2000, which is incorporated by reference herein in its entirety.

1. INTRODUCTION The present invention relates to compositions and methods of preparing immunogenic material that increases a subject's immune response to a vaccine for the prevention or treatment of cancer or infectious diseases. Heat shock proteins (HSPs) including, but not limited to, hsp70, hsp90 and gp96 alone or in combination with each other are administered in conjunction with a vaccine to augment the immune response of a subject against tumors and infectious agents. The present invention also contemplates administration of alpha (2) macroglobulin (o2M) in conjunction with a vaccine to augment the immune response of a subject against tumors and infectious agents.

2. BACKGROUND OF THE INVENTION Citation or discussion of a reference herein shall not be construed as an admission that such is prior art to the present invention.

2.1 Vaccines Vaccination has eradicated certain diseases such as polio, tetanus, chicken pox, and measles in many countries. This approach has exploited the ability of the immune system to resist and prevent infectious diseases.

Traditional ways of preparing vaccines include the use of inactivated or attenuated pathogens. A suitable inactivation of the pathogenic microorganism renders it harmless as a biological agent but does not destroy its immunogenicity. Injection of these "killed"particles into a host will then elicit an immune response capable of preventing a future infection with a live microorganism. However, a major concern in the use of inactivated pathogens as vaccines is the failure to inactivate all the microorganisms. Even when this is accomplished, since killed pathogens do not multiply in their host, or for other unknown reasons, the immunity achieved is often incomplete, short lived and requires

multiple immunizations. Finally, the inactivation process may alter the microorganism's antigens, rendering them less effective as immunogens.

Attenuation refers to the production of strains of pathogenic microorganisms which have essentially lost their disease-producing ability. One way to accomplish this is to subject the microorganism to unusual growth conditions and/or frequent passage in cell culture. Mutants are then selected which have lost virulence but yet are capable of eliciting an immune response. Attenuated pathogens often make good immunogens as they actually replicate in the host cell and elicit long lasting immunity. However, several problems are encountered with the use of live vaccines, the most worrisome being insufficient attenuation and the risk of reversion to virulence.

An alternative to the above methods is the use of subunit vaccines. This involves immunization only with those components which contain the relevant immunological material. A new promising alternative is the use of DNA or RNA as vaccines. Such genetic vaccines have progressed from an idea to entities being studied in clinical trials (See, Weiner and Kennedy, July 1999, Scientific American, pp. 50-57).

Vaccines are often formulated and inoculated with various adjuvants. The adjuvants aid in attaining a more durable and higher level of immunity using small amounts of antigen or fewer doses than if the immunogen were administered alone. The mechanism of adjuvant action is unpredictable, complex and not completely understood (See Suzue, et al. , 1996, Basel: Birkhauser Verlag, 454-55).

Because of the risks associated with inactivated and attenuated pathogens, the ability to boost or amplify an immune response to minimal quantities of a vaccine would be ideal and advantageous. Furthermore, as the mechanism of adjuvants is not completely understood and is still unpredictable, alternative methods of boosting a subject's immune response with current methods of vaccination is highly desirable.

2.2 Immune Responses An organism's immune system reacts with two types of responses to pathogens or other harmful agents-humoral response and cell-mediated response (See <BR> <BR> Alberts, B. et al. , 1994, Molecular Biology of the Cell. 1195-96). When resting B cells are activated by antigen to proliferate and mature into antibody-secreting cells, they produce and secrete antibodies with a unique antigen-binding site. This antibody-secreting reaction is known as the humoral response. On the other hand, the diverse responses of T cells are

collectively called cell-mediated immune reactions. There are two main classes of T cells- cytotoxic T cells and helper T cells. Cytotoxic T cells directly kill cells that are infected with a virus or some other intracellular microorganism. Helper T cells, by contrast, help stimulate the responses of other cells: they help activate macrophages, dendritic cells and B <BR> <BR> cells, for example (See Alberts, B. et al. , 1994, Molecular Biology of the Cell. 1228). Both cytotoxic T cells and helper T cells recognize antigen in the form of peptide fragments that are generated by the degradation of foreign protein antigens inside the target cell, and both, therefore, depend on major histocompatibility complex (MHC) molecules, which bind these peptide fragments, carry them to the cell surface, and present them there to the T cells (See Alberts, B. et al. , 1994, Molecular Biology of the Cell. 1228). MHC molecules are typically found in abundance on antigen-presenting cells (APCs).

2.3 Antigen Presentation Antigen-presenting cells (APCs), such as macrophages and dendritic cells, are key components of innate and adaptive immune responses. Antigens are generally 'presented'to T cells or B cells on the surfaces of other cells, the APCs. APCs can trap lymph-and blood-borne antigens and, after internalization and degradation, present antigenic peptide fragments, bound to cell-surface molecules of the major histocompatibility complex (MHC), to T cells. APCs may then activate T cells (cell- mediated response) to clonal expansion, and these daughter cells may either develop into cytotoxic T cells or helper T cells, which in turn activate B (humoral response) cells with the same MHC-bound antigen to clonal expansion and specific antibody production (See Alberts, B. et al. , 1994, Molecular Biology of the Cell. 1238-45).

Two types of antigen-processing mechanisms have been recognized. The first type involves uptake of proteins through endocytosis by APCs, antigen fragmentation within vesicles, association with class II MHC molecules and expression on the cell surface.

This complex is recognized by helper T cells expressing CD4. The other is employed for proteins, such as viral antigens, that are synthesized within the cell and appears to involve protein fragmentation in the cytoplasm. Peptides produced in this manner become associated with class I MHC molecules and are recognized by cytotoxic T cells expressing CD8 (See Alberts, B. et al. , 1994, Molecular Biology of the Cell. 1233-34).

Stimulation of T cells involves a number of accessory molecules expressed by both T cell and APC. Co-stimulatory molecules are those accessory molecules that

promote the growth and activation of the T cell. Upon stimulation, co-stimulatory molecules induce release of cytokines, such as interleukin 1 (IL-1) or interleukin 2 (IL-2), interferon, etc. , which promote T cell growth and expression of surface receptors (See Paul, 1989, Fundamental Immunology. 109-10).

Normally, APCs are quiescent and require activation for their function. The identity of signals which activate APCs is a crucial and unresolved question (See Banchereau, et al. , 1998, Nature 392: 245-252; Medzhitov, et al. , 1998, Curr Opin Immunol.

10: 12-15).

2.4 Heat Shock Proteins Heat shock proteins, also known as stress proteins, are intracellular molecules that are abundant, soluble, and highly conserved. As intracellular chaperones, HSPs participate in many biochemical pathways of protein maturation and function active during times of stress and normal cellular homeostasis. Many stresses can disrupt the three- dimensional structure, or folding, of a cell's proteins. Left uncorrected, mis-folded proteins form aggregates that may eventually kill the cell. HSPs bind to those damaged proteins, helping them refold into their proper conformations. In normal (unstressed) cellular homeostasis, HSPs are required for cellular metabolism. HSPs help newly synthesized polypeptides fold and thus prevent premature interactions with other proteins. Also, HSPs aid in the transport of proteins throughout the cell's various compartments.

The major HSPs can accumulate to very high levels in stressed cells, but they occur at low to moderate levels in cells that have not been stressed. For example, the highly inducible mammalian hsp70 is hardly detectable at normal temperatures but becomes one of the most actively synthesized proteins in the cell upon heat shock (Welch et al., 1985, J. Cell. Biol. 101: 1198-1211). In contrast, hsp90 and hsp60 proteins are abundant at normal temperatures in most, but not all, mammalian cells and are further induced by heat (Lai et al. , 1984, Mol. Cell. Biol. 4: 2802-2810 ; van Bergen en Henegouwen et al. , 1987, Genes Dev. 1: 525-531).

HSPs have been found to have immunological and antigenic properties.

Immunization of mice with gp96 or p84/86 isolated from a particular tumor rendered the mice immune to that particular tumor, but not to antigenically distinct tumors (Srivastava, P. K. et al. , 1988, Immunogenetics 28: 205-207; Srivastava, P. K. et al. , 1991, Curr. Top.

Microbiol. Immunol. 167: 109-123). Further, hsp70 was shown to elicit immunity to the

tumor from which it was isolated but not to antigenically distinct tumors. However, hsp70 <BR> <BR> depleted of peptides was found to lose its specific immunogenic activity (Udono, M. , and<BR> Srivastava, P. K. , 1993, J. Exp. Med. 178: 1391-1396). These observations suggested that the heat shock proteins are not antigenic per se, but form noncovalent complexes with antigenic peptides, and the complexes can elicit specific immunity to the antigenic peptides <BR> <BR> (Srivastava, P. K. , 1993, Adv. Cancer Res. 62: 153-177; Udono, H. et al. , 1994, J. Immunol.,<BR> 152: 5398-5403; Suto, R. et al. , 1995, Science, 269: 1585-1588). Recently, hsp60 and hsp70 have been found to stimulate production of proinflammatory cytokines, such as TNFa and <BR> <BR> IL-6, by monocytes, macrophages, or cytotoxtic T cells (Breloer et al. , 1999, J. Immunol.<BR> <P>162: 3141-3147; Chen et al. , 1999, J. Immunol. 162: 3212-3219; Ohashi et al. , 2000, J.<BR> <P>Immunol. 164: 558-561; Asea et al. , 2000, Nature Medicine, 6: 435-442; Todryk et al. , 1999, J. Immunol. 163: 1398-1408). Hsp70 has also been shown to target immature dendritic cells <BR> <BR> and make them more able to capture antigens (Todryk et al. , J. Immunol. 163: 1398-1408).

It has been postulated that release of or induction of expression of hsp60 and hsp70, e. g., due to cell death, may serve to signal that an immune reaction should be raised (Chen et al., <BR> <BR> 1999, J. Immunol. 162: 3212-3219; Ohashi et al. , 2000, J. Immunol. 164: 558-561; Todryk et<BR> al. , 1999, J. Immunol. 163: 1398-1408; Basu et al. Intl. Immunol. 2000 vol 12: 1539-1546).

The use of noncovalent complexes of HSP and peptide, purified from cancer cells, for the treatment and prevention of cancer has been described in U. S. Patent Nos.

5,750, 119,5, 837,251, and 6,017, 540.

The use of HSP-peptide complexes for sensitizing antigen presenting cells in vitro for use in adoptive immunotherapy is described in United States Patent Nos. 5,985, 270 and 5,830, 464.

HSP-peptide complexes can also be isolated from pathogen-infected cells and used for the treatment and prevention of infection caused by the pathogen, such as viruses, and other intracellular pathogens, including bacteria, protozoa, fungi and parasites; see United States Patent Nos. 5,961, 979, and 6,048, 530.

Immunogenic HSP-peptide complexes can also be prepared by in vitro complexing of HSPs and antigenic peptides, and the uses of such complexes for the treatment and prevention of cancer and infectious diseases has been described in United States Patent Nos. 5,935, 576, and 6,030, 618. The use of heat shock protein in combination

with a defined antigen for the treatment of cancer and infectious diseases have also been described in PCT publication W097/06821 dated February 27,1997.

The purification of HSP-peptide complexes from cell lysate has been described previously; see for example, United States Patent Nos. 5,750, 119, and 5,997, 873 ; 2.5 o2-Macroglobulin The a-macroglobulins are members of a protein superfamily of structurally related proteins which also comprises complement components C3, C4 and C5. The human plasma protein alpha (2) macroglobulin (a2M) is a 720 kDa homotetrameric protein primarily known as proteinase inhibitor and plasma and inflammatory fluid proteinase scavenger molecule (for review see Chu and Pizzo, 1994, Lab. Invest. 71: 792). Alpha (2) macroglobulin is synthesized as a 1474 amino acid precursor, the first 23 of which function as a signal sequence that is cleaved to yield a 1451 amino acid mature protein (Kan et al., 1985, Proc. Natl. Acad. Sci. U. S. A. 82: 2282-2286).

Alpha (2) macroglobulin promiscuously binds to proteins and peptides with nucleophilic amino acid side chains in a covalent manner (Chu et al., 1994, Ann. N. Y.

Acad. Sci. 737: 291-307) and targets them to cells which express the c2M receptor (o2MR) (Chu and Pizzo, 1993, J. Immunol. 150: 48). Binding of c2M to the o2MR is mediated by the C-terminal portion of c2M (Holtet et al., 1994, FEBS Lett. 344: 242-246) and key residues have been identified (Nielsen et al., 1996, J. Biol. Chem. 271: 12909-12912).

Generally known for inhibiting protease activity, o2M binds to a variety of proteases thorough multiple binding sites (see, e. g., Hall et al., 1981, Biochem. Biophys.

Res. Commun. 100 (1) : 8-16). Protease interaction with o2M results in a complex structural rearrangement called transformation, which is the result of a cleavage within the"bait" region of o2M after the proteinase becomes"trapped"by thioesters. The conformational change exposes residues required for receptor binding, allowing the oc2M-proteinase complex to bind to the o2MR. Methylamine can induce similar conformational changes and cleavage as that induced by proteinases. The cleaved form of c2M, which is not recognized by the receptor, is often referred to as the"slow"form (s-c2M). The cleaved form is referred to as the"fast"form (f-oe2M) (reviewed by Chu et al., 1994, Ann. N. Y.

Acad. Sci. 737: 291-307).

Studies have shown that, in addition to its proteinase-inhibitory functions, a2M, when complexed to antigens, can enhance the antigens'ability to be taken up by

antigen presenting cells such as macrophages and presented to T cell hybridomas in vitro by up to two orders of magnitude (Chu and Pizzo, 1994, Lab. Invest. 71: 792), and induce T cell proliferation (Osada et al., 1987, Biochem. Biophys. Res. Commun. 146 : 26-31).

Further evidence suggests that complexing antigen with cOM enhances antibody production by crude spleen cells in vitro (Osada et al., 1988, Biochem. Biophys. Res. Commun.

150: 883) and elicits an in vivo antibody responses in experimental rabbits (Chu et al., 1994, J. Immunol. 152: 1538-1545) andmice (Mitsuda et al., 1993, Biochem. Biophys. Res.

Commun. 101 : 1326-1331). However, none of these studies have shown whether o2M- antigen complexes are capable of eliciting cytotoxic T cell responses in vivo. o2M can form complexes with antigens, which are taken up by antigen presenting cells ("APCs") via the o2MR, also known as LDL (low-density lipoprotein) Receptor-Related Protein ("LRP") or CD91 (see PCT/US01/18047, which is incorporated by reference herein in its entirety). o2M directly competes for the binding of heat shock protein gp96 to the a2MR, indicating that oe2M and HSPs may bind to a common recognition site on the o2MR (Binder et al., 2000, Nature Immunology 1 (2), 151-154).

Additionally, o2M-antigenic peptide complexes prepared in vitro can be administered to animals to generate a cytotoxic T cell response specific to the antigenic molecules (Binder et al., 2001, J. Immunol. 166: 4968-72). Thus, because HSPs and o2M have a number of common functional attributes, such as the ability to bind peptide, the recognition and uptake by the od2MR, and the stimulation of a cytotoxic T cell response, o2M can be used for immunotherapy against cancer and infectious disease.

3. SUMMARY OF THE INVENTION The present invention provides for a method of producing or increasing an immune response elicited by vaccines using HSPs or a2M. The source of the HSP or o2M is preferably an eukaryote, and most preferably a mammal.

In one embodiment of the invention, the method for producing an immune response comprises administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced; and administering to the subject a heat shock protein preparation, wherein the immune response against the component is not elicited in the absence of the administering of the vaccine composition.

The heat shock protein preparation does not display the immunogenicity of the component.

The heat shock protein preparation alone cannot elicit an immune response against the

component in the absence of the administering of the vaccine composition. The method can increase the magnitude of the immune response to the component of interest relative to that obtained in the absence of administering to the subject a heat shock protein preparation. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an 02M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. hi another preferred embodiment, if the component of the vaccine composition is a peptide complexed to oe2M, the vaccine composition and the o2M preparation are not present in admixture.

In one embodiment of the invention, the method for producing an immune response comprises administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced; and administering to the subject an c2M preparation, wherein the immune response against the component is not elicited in the absence of the administering of the vaccine composition. The o2M preparation does not display the immunogenicity of the component. The o2M preparation alone cannot elicit an immune response against the component in the absence of the administering of the vaccine composition. The method can increase the magnitude of the immune response to the component of interest relative to that obtained in the absence of administering to the subject an o2M preparation. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to o2M, the vaccine composition and the o2M preparation are not present in admixture.

In another embodiment, the invention provides for a method of inducing an immune response by a sub-immunogenic amount of a vaccine composition, wherein the HSP preparation facilitates the induction of an immune response by an amount of vaccine composition which is otherwise insufficient for inducing the immune response when used alone. In particular, the method comprises the steps of (a) administering to the subject an amount of a heat shock protein preparation; and (b) administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced in an amount that is sub-immunogenic in the absence of step (a), whereby an immune response to said component is induced in the subject, and wherein the heat shock protein preparation does not display the immunogenicity of the component. The heat shock protein preparation does not elicit an immune response against said component in the absence of said administering of the vaccine composition. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to ol2M, the vaccine composition and the a2M preparation are not present in admixture.

In another embodiment, the invention provides for a method of inducing an immune response by a sub-immunogenic amount of a vaccine composition, wherein the o2M preparation facilitates the induction of an immune response by an amount of vaccine composition which is otherwise insufficient for inducing the immune response when used alone. In particular, the method comprises the steps of (a) administering to the subject an amount of a o2M preparation ; and (b) administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced in an amount that is sub-immunogenic in the absence of step (a), whereby an immune response to said component is induced in the subject, and wherein the odM preparation does not display the immunogenicity of the component. The o2M preparation does not elicit an immune response against said component in the absence of said administering of the

vaccine composition. In a preferred embodiment, the vaccine composition is not an HSP- peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to o2M, the vaccine composition and the o2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of treating or preventing an infectious disease in a subject comprising administering to the subject a vaccine composition comprising a component that displays the antigenicity of an infectious agent that causes the infectious disease; and administering to the subject an amount of a heat shock protein preparation effective in combination with the vaccine composition to induce or increase an immune response to the component in the subject. The heat shock protein preparation does not display the immunogenicity of the component. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an a2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an c2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to o2M, the vaccine composition and the o2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of treating or preventing an infectious disease in a subject comprising administering to the subject a vaccine composition comprising a component that displays the antigenicity of an infectious agent that causes the infectious disease; and administering to the subject an amount of a o2M preparation effective in combination with the vaccine composition to induce or increase an immune response to the component in the subject. The o2M preparation does not display the immunogenicity of the component. In a preferred embodiment, the vaccine

composition is not an HSP-peptide complex. hi another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an a2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to o2M, the vaccine composition and the o2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of treating or preventing a cancer in a subject comprising administering to the subject a vaccine composition comprising a component that displays the antigenicity of a cancer cell; and administering to the subject an amount of a heat shock protein preparation effective to induce or increase an immune response in the subject to the component wherein the heat shock protein preparation does not display the immunogenicity of the component. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an a2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to a2M, the vaccine composition and the o2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of treating or preventing a cancer in a subject comprising administering to the subject a vaccine composition comprising a component that displays the antigenicity of a cancer cell; and administering to the subject an amount of a a2M preparation effective to induce or increase an immune response in the subject to the component wherein the o2M preparation does not display the immunogenicity of the component. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine

composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an a 2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to a2M, the vaccine composition and the o2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of inducing an immune response by a vaccine composition in a subject comprising administering to the subject a heat shock protein preparation; and administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced, the vaccine composition being in an amount that is sub-immunogenic for the component in the absence of the vaccine composition. The heat shock protein preparation does not display the immunogenicity of the component. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an a2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to o2M, the vaccine composition and the c2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of inducing an immune response by a vaccine composition in a subject comprising administering to the subject a o2M preparation; and administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced, the vaccine composition being in an amount that is sub-immunogenic for the component in the absence of the vaccine composition. The o2M preparation does not display the immunogenicity of the component. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine composition

does not comprise a heat shock protein. In another preferred embodiment, the vaccine composition does not comprise an oe2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to o2M, the vaccine composition and the a2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of activating antigen presenting cells comprising contacting APCs with a heat shock protein preparation.

In particular, the antigen presenting cells can be obtained from an individual, the APCs being optionally expanded and/or purified, and treated ex vivo with a heat shock protein preparation. The treated APCs can then be administered to a subject concurrently, before, or after with the administration of a vaccine composition against which an immune response is desired to be induced. The patient may be treated according to the present invention with a vaccine composition and with activated APCs and/or an HSP preparation. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex.

In yet another embodiment, the invention provides a method of activating antigen presenting cells comprising contacting APCs with a o2M preparation. In particular, the antigen presenting cells can be obtained from an individual, the APCs being optionally expanded and/or purified, and treated ex vivo with a a2M preparation. The treated APCs can then be administered to a subject concurrently, before, or after with the administration of a vaccine composition against which an immune response is desired to be induced. The patient may be treated according to the present invention with a vaccine composition and with activated APCs and/or an HSP preparation. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex.

In these above-mentioned embodiments of the invention, the heat shock protein preparation and the o2M preparation do not elicit an immune response against the component in the absence of the administration of the vaccine composition. The heat shock protein preparation and the o2M preparation do not display the immunogenicity of the component in the vaccine composition. The immunogenicity of a heat shock protein

preparation or a o2M preparation can be tested in vivo or in vitro by any method known in the art, such as but not limited to those described in section 5.5.

In various embodiments, the HSPs or ot2M are administered into a subject before the administration of a vaccine composition. Alternatively, the HSPs or a2M are administered to the subject concurrently with the administration of a vaccine composition, preferably a vaccine that is not a hsp-peptide complex. The HSPs or a2M can also be administered to the subject after the administration of a vaccine composition. Preferably, the subject is mammalian, or, more specifically, human.

The present invention further provides a method for improving the outcome of a treatment in a subject receiving a therapeutic modality which is not a vaccine. The method comprises administering a mammalian heat shock protein preparation or a o2M preparation to the subject before, concurrently with, or after the administration of the therapeutic modality.

Without being bound by any theory, an increased concentration of HSP induces secretion of cytokines and surface expression of antigen-presenting and co- stimulatory molecules. It is also believed that an increased concentration of Q2M induces secretion of cytokines and surface expression of antigen-presenting and co-stimulatory molecules. Applicant's experimentation with CD1 lb+ cell activation shows that the presence of HSPs in the extracellular milieu induces interleukin-lß secretion and surface expression of MHC class II molecules. The activation of APCs increases the affinity between the resultant antigen-MHC complexes from the vaccine and T-cell antigen surface receptors (TCRs) on the surface of the T-cells. Accordingly, the HSP preparation administered to a subject can boost the effectiveness of a vaccine by increasing the efficiency and effectiveness of antigen presentation.

The HSP preparation used in the methods of the invention can include free HSP not bound to any molecule, and molecular complexes of HSP with another molecule, such as a peptide. An HSP-peptide complex comprises a HSP covalently or noncovalently attached to a peptide. The methods of the invention do not require covalent or noncovalent attachment of an HSP to any specific antigens or antigenic peptides prior to administration to a subject.

The o2M preparation used in the methods of the invention can include free o2M not bound to any molecule, and molecular complexes of c2M with another molecule,

such as a peptide. An o2M-peptide complex comprises a o2M covalently or noncovalently attached to a peptide. The methods of the invention do not require covalent or noncovalent attachment of an o2M to any specific antigens or antigenic peptides prior to administration to a subject.

Also encompassed in the invention are kits comprising one or more containers each containing a heat shock protein preparation in an amount effective to increase an immune response elicited by a vaccine composition against a component of the vaccine composition against which an immune response is desired; and one or more containers each containing the vaccine composition in an amount that, when administered before, concurrently with, or after the administration of the heat shock protein preparation of (a), is effective to induce an immune response against the component. The invention also encompasses kits comprising one or more containers each containing a o2M preparation in an amount effective to increase an immune response elicited by a vaccine composition against a component of the vaccine composition against which an immune response is desired; and one or more containers each containing the vaccine composition in an amount that, when administered before, concurrently with, or after the administration of the o2M preparation of (a), is effective to induce an immune response against the component. In other embodiments, the invention provides kits comprising one or more containers each containing a a2M preparation in an amount effective to increase an immune response elicited by a vaccine composition against a component of the vaccine composition against which an immune response is desired; and one or more containers each containing the vaccine composition in an amount that, when administered before, concurrently with, or after the administration of the o2M preparation of (a), is effective to induce an immune response against the component. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In a preferred embodiment, the vaccine composition is not an oe2M-peptide complex.

4. BRIEF DESCRIPTION OF THE FIGURES Figure 1A-1D. Figure 1A. SDS-PAGE analysis of purified preparations of gp96, hsp90 and hsp70. The HSPs were purified from livers of C57BL/6 mice, as described in Section 6. 1. Two, ug of each HSP preparation was applied to each lane. Figure 1B.

Peritoneal cells obtained from C57BL/6 mice injected intraperitoneally with pristane were positively selected for CDl lb+ cells. Cells (5x104) were incubated for 20 hours at 37°C in

complete RPMI medium with 5% fetal calf serum alone, or with increasing quantities of homogenous preparation of gp96 (Figure 1B) or hsp90 or hsp70 (Figure 1C) purified from livers of C57BL/6 mice, as indicated, in the same medium. Supernatants were harvested and assayed by ELISA for TNF-x, IL-12, IL-l# and BM-CSF. Cultures of CDl lb+ cells were also similarly incubated with non-HSPs such as histone, ovalbumin and insulin and the supernatants tested for TNF-oe (Figure 1D).

Figure 2. The APC-stimulating activity of gp96 is abridged in an LPS- hyporesponsive mouse strain. CD1 lb+ cells (5x104), isolated from C3H/HeN or C3H/HeJ strains of mice as described in Section 6.1, were incubated in complete RPMI medium with 5% fetal calf serum alone, or treated with gp96 at the indicated amounts in the same medium for 20 hrs at 37°C. Supernatants were harvested and assayed for IL-lß and TNF-oe as indicated, by ELISA.

Figure 3A-3C. The APC-stimulating activity of gp96 does not derive from contaminating LPS. Figure 3A. CD1 lb+ cells (5x104), isolated from C57BL/6 mice as described were incubated in complete RPMI medium with 5% fetal calf serum alone, or treated with gp96, LPS or BSA at the indicated amounts in the same medium for 20 hrs at 37°C. Supernatants were harvested and assayed for IL-lß and TNF-o, as indicated by ELISA. Figure 3B. CD1 lb+ cells (5x104), isolated from C57BL/6 mice were incubated in complete RPMI medium with or without 5% fetal calf serum (as a source of LBP) as indicated, or treated with gp96 or LPS at the indicated amounts in the above media for 20 hrs at 37°C. Supernatants were harvested and assayed for IL-10 by ELISA. Figure 3C.

The LPS antagonist Rslp, derived from Rhodopseudomonas spheroides (2yg/ml) was added to cytokine secretion assay of LPS (2, ltg/ml) or gp96 (90yg/ml) as indicated.

Figure 4. HSPs stimulate CDl lc+ cells to express antigen presenting and co- stimulatory molecules. Bone marrow-derived DC cultures were exposed to the medium, HSPs (400, ug/ml) or LPS (2 pg/ml) for 20 hours, harvested and analyzed for expression of the cell surface molecules indicated. GM-CSF was not present in the DC cultures when they were treated with medium alone, or gp96, or LPS or albumin. The percentages shown are CD lc+ cells that are also positive for the indicated surface markers. Cells were analyzed by flow cytometry using the FACScan (Becton Dickinson, La Jolla, California).

Live cells were gated based on FSC/SSC profiles.

Figure 5. Gp96 interacts with APCs through the NFKB signal transduction pathway. (A) DCs (1X106 cells) were pulsed with gp96 (100 ug/ml) or LPS (4 llg/ml) for the indicated time points. Nuclear extracts of unpulsed or pulsed cultures were prepared and were used in binding to NFKB-specific oligomer as described in Methods. The complexes were resolved by native PAGE and autoradiographed. (B) The data from (A) are quantitated by scanning the gels under linear conditions of exposure, and plotted.

Figure 6A-6B. Exposure of DCs to necrotic but not apoptotic cells leads to maturation of DCs and to nuclear translocation of NFKB. (A) Cultures of immature DCs (2 x 106) were pulsed with medium alone, or 106 cell equivalents each of necrotic or apoptotic E. G7 cells, or LPS (as a positive control) for 20 h. DC cultures were monitored, for expression of surface markers as indicated. (B) DC cultures exposed to medium alone, or to necrotic or apoptotic E. G7 cells for 15 minutes and were analyzed for translocation of NFKB as described in legend to Fig. 5.

5. DETAILED DESCRIPTION OF THE INVENTION The present invention provides a method of producing or increasing an immune response elicited by a vaccine composition, comprising administering heat shock proteins (HSPs) or a2-macroglobulin (o2M) in conjunction with the administration of the vaccine composition.

Some of the current vaccination strategies use attenuated viral and bacterial strains or whole cells that have been killed to induce an immune response in a subject in whom treatment or prevention of an infectious disease or cancer is desired. However, these strategies carry the risk that the attenuated strains may recombine genetically with the host DNA and turn into a virulent strain. Thus, the ability to boost or increase an immune response using the claimed methods with these vaccines is desirable and advantageous.

Additionally, the ability to augment or amplify a subject's immune response using the claimed method with a generally weak vaccine presents a safer and more feasible alternative to using larger dosages of the weak vaccine. The methods of the invention can also aid the induction of an immune response by an amount of vaccine composition that is insufficient to induce an immune response if used alone. The methods of the invention can be used with any type of vaccine composition comprising a component against which an immune response is desired, including but not limited to, live vaccine, attenuated vaccine, subunit vaccine, DNA vaccine, and RNA vaccine. In a preferred embodiment, the vaccine

composition is not an HSP-peptide complex vaccine. In another preferred embodiment, the vaccine composition is not an ou2M-peptide complex vaccine. In another preferred embodiment, the vaccine composition does not comprise a HSP or a a2M. The vaccine composition may comprise an adjuvant. The vaccine composition may be administered with one or more adjuvants.

In the present invention, an HSP preparation or an o2M preparation is administered to a subject, preferably at a site where APCs are expected to encounter the antigen (s) (molecular components against which an immune response is desired to be induced) in a vaccine composition, before, concurrently with, or after the administration of the vaccine composition. The HSP preparations and o2M preparations of the invention activate APCs and thus, increase the effectiveness and/or the efficiency of antigen presentation. Accordingly, the present invention provides for a method of using an HSP preparation to increase a subject's immune response elicited by the vaccine composition.

The invention also provides for a method of using an o2M preparation to increase a subject's immune response elicited by the vaccine composition. The activation of APCs by the HSP preparations or by the oe2M preparations ex vivo and the subsequent administration of activated APCs are encompassed in the present invention. Such administration of activated APCs can be carried out, before, concurrently with, or after the administration of a vaccine composition, which vaccine composition, and activated APCs, may be administered before, concurrently with, or after the administration of a HSP preparation or alternatively a o2M preparation, according to the methods of the invention. Thus, a patient may be treated according to the present invention with a vaccine composition and with activated APCs and/or an HSP preparation. A HSP preparation that is the same as or different from the HSP preparation to be administered can be used for activating the APCs. A patient may also be treated according to the present invention with a vaccine composition and with activated APCs and/or an o2M preparation. A c2M preparation that is the same as or different from the o2M preparation to be administered can be used for activating the APCs.

Without being bound by any theory or mechanism, the applicants believe that the HSP preparation or the o2M preparation, upon contact with APCs at a site, upregulates expression of co-stimulatory molecules on the cell surface of APCs, and increases cytokine production. Although not limited to this mechanism, increase or amplification of a subject's immune response is likely induced by the upregulation of co-

stimulatory molecules and other molecules required for antigen presentation on the APCs, such as B7-1, B7-2, and MHC class II, and their ensuing increase in production of cytokines, soluble molecules that mediate interaction between cells, often promoting immune cell growth and division. Because of this HSP-induced or o2M-induced stimulation of co-stimulatory molecules, the claimed methods generally boost T-cell activation and increase a subject's immune response. As a result, an increased number of activated APCs are available to present to T-cells antigens, including those present in a vaccine administered in the same immunological time frame. The ability of HSPs and oe2M to activate APCs can confer a distinct immunological advantage to the subject. However, it should be noted that the present invention is not to be limited in scope by the mechanism described herein.

For the purposes of this invention, an HSP preparation is a composition <BR> <BR> comprising HSPs whether unbound or bound to other molecules (e. g. , peptides). The HSPS are preferably purified. An HSP preparation may include crude cell lysate comprising HSP, the amount of lysate corresponding to between 100 to 108 cell equivalents. When a peptide is attached to a HSP, the peptide may be any peptide, which can be noncovalently or covalently bound to the HSP. An o2M preparation is a composition comprising a2M whether unbound or bound to other molecules (e. g. , peptides). The 02M is preferably purified. An o2M preparation may include crude cell lysate comprising o2M, the amount of lysate corresponding to between 100 to 108 cell equivalents. When a peptide is attached to a a2M, the peptide may be any peptide, which can be noncovalently or covalently bound to the o2M. HSPs can be conveniently purified from most cellular sources as a population of complexes of different peptides non-covalently bound to HSPs. Similarly, o2M can be conveniently purified from most cellular sources as a population of complexes of different peptides non-covalently bound to a2M. The peptide (s) may be unrelated to the vaccine composition, or the infectious disease or disorder in question. The HSPs or c2M can be separated from the non-covalently bound peptides by exposure to low pH and/or adenosine triphosphate, or other methods known in the art.

Generally, the HSP preparation or o2M preparation is separately administered from the vaccine composition. For convenience and comfort of a recipient, the HSP preparation or the o2M preparation can be mixed with the vaccine composition immediately prior to administration. When the HSP preparation or o2M preparation is not

used in conjunction with a vaccine composition to elicit a specific immune response, administering the HSP preparation or a2M preparation, respectively, alone does not induce the antigen-specific immune response that would have been induced by the vaccine composition. When administering a single composition comprising both the vaccine composition and HSP preparation, the vaccine composition is preferably not an HSP- peptide complex vaccine. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. When administering a single composition comprising both the vaccine composition and o2M preparation, the vaccine composition is preferably not an oe2M-peptide complex vaccine. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the o2M preparation are not present in admixture.

In various embodiments, the source of the HSP or cm is preferably an eukaryote, more preferably a mammal, and most preferably a human. Accordingly, the HSP preparation used by the methods of the invention includes eukaryotic HSPs, mammalian HSPs and human HSPs. The eukaryotic source from which the HSP preparation is derived and the subject receiving the HSP preparation are preferably the same species. Similarly, the a2M preparation used by the methods of the invention includes eukaryotic o2M, mammalian o2M and human a2M. The eukaryotic source from which the a2M preparation is derived and the subject receiving the o2M preparation are preferably the same species.

The HSP preparation or the o2M preparation can be administered prior to, concurrently with, or subsequent to the administration of a vaccine composition.

In one embodiment, the HSP preparation is administered to a subject at reasonably the same time as the vaccine. This method provides that the two administrations are performed within a time frame of less than one minute to about five minutes, or up to about sixty minutes from each other, for example, at the same doctor's visit.

In one embodiment, the a2M preparation is administered to a subject at reasonably the same time as the vaccine. This method provides that the two administrations are performed within a time frame of less than one minute to about five minutes, or up to about sixty minutes from each other, for example, at the same doctor's visit.

In one embodiment, the HSP preparation and vaccine composition are administered at exactly the same time. In another embodiment the HSP preparation and vaccine composition are administered in a sequence and within a time interval such that the HSP preparation and vaccine composition can act together to provide an increased benefit than if they were administered alone. In another embodiment, the HSP preparation and a vaccine composition are administered sufficiently close in time so as to provide the desired therapeutic or prophylactic outcome. Each can be administered simultaneously or separately, in any appropriate form and by any suitable route. In one embodiment, the HSP preparation and vaccine composition are administered by different routes of administration.

In an alternate embodiment, each is administered by the same route of administration. The HSP preparation can be administered at the same or different sites, e. g. arm and leg.

Preferably, when administered simultaneously, the HSP preparation and the vaccine composition are not administered in admixture or at the same site of administration by the same route of administration.

In one embodiment, the o2M preparation and vaccine composition are administered at exactly the same time. In another embodiment the c2M preparation and vaccine composition are administered in a sequence and within a time interval such that the o2M preparation and vaccine composition can act together to provide an increased benefit than if they were administered alone. In another embodiment, the o2M preparation and a vaccine composition are administered sufficiently close in time so as to provide the desired therapeutic or prophylactic outcome. Each can be administered simultaneously or separately, in any appropriate form and by any suitable route. In one embodiment, the o2M preparation and vaccine composition are administered by different routes of administration.

In an alternate embodiment, each is administered by the same route of administration. The o2M preparation can be administered at the same or different sites, e. g. arm and leg.

Preferably, when administered simultaneously, the o2M preparation and the vaccine composition are not administered in admixture or at the same site of administration by the same route of administration.

In various embodiments, the HSP preparation and vaccine composition are administered less than 1 hour apart, at about 1 hour apart, 1 hour to 2 hours apart, 2 hours to 3 hours apart, 3 hours to 4 hours apart, 4 hours to 5 hours apart, 5 hours to 6 hours apart, 6 hours to 7 hours apart, 7 hours to 8 hours apart, 8 hours to 9 hours apart, 9 hours to 10 hours

apart, 10 hours to 11 hours apart, 11 hours to 12 hours apart, no more than 24 hours apart or no more than 48 hours apart. In other embodiments, the HSP preparation and vaccine composition are administered 2 to 4 days apart, 4 to 6 days apart, 1 week a part, 1 to 2 weeks apart, 2 to 4 weeks apart, one moth apart, 1 to 2 months apart, or 2 or more months apart. In preferred embodiments, the HSP preparation and vaccine composition are administered in a time frame where both are still active. One skilled in the art would be able to determine such a time frame by determining the half life of each administered component.

In various embodiments, the o2M preparation and vaccine composition are administered less than 1 hour apart, at about 1 hour apart, 1 hour to 2 hours apart, 2 hours to 3 hours apart, 3 hours to 4 hours apart, 4 hours to 5 hours apart, 5 hours to 6 hours apart, 6 hours to 7 hours apart, 7 hours to 8 hours apart, 8 hours to 9 hours apart, 9 hours to 10 hours apart, 10 hours to 11 hours apart, 11 hours to 12 hours apart, no more than 24 hours apart or no more than 48 hours apart. In other embodiments, the o2M preparation and vaccine composition are administered 2 to 4 days apart, 4 to 6 days apart, 1 week a part, 1 to 2 weeks apart, 2 to 4 weeks apart, one moth apart, 1 to 2 months apart, or 2 or more months apart. In preferred embodiments, the Q2M preparation and vaccine composition are administered in a time frame where both are still active. One skilled in the art would be able to determine such a time frame by determining the half life of each administered component.

In one embodiment, the HSP preparation and vaccine composition are administered within the same patient visit. In a specific preferred embodiment, the HSP preparation is administered prior to the administration of the vaccine composition. In an alternate specific embodiment, the HSP preparation is administered subsequent to the administration of the vaccine composition.

In one embodiment, the o2M preparation and vaccine composition are administered within the same patient visit. In a specific preferred embodiment, the o2M preparation is administered prior to the administration of the vaccine composition. In an alternate specific embodiment, the o2M preparation is administered subsequent to the administration of the vaccine composition.

In certain embodiments, the HSP preparation or the o2M preparation and vaccine composition are cyclically administered to a subject. Cycling therapy involves the

administration of the HSP preparation or an a2M preparation for a period of time, followed by the administration of a vaccine composition for a period of time and repeating this sequential administration. Cycling therapy can reduce the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improve the efficacy of the treatment. In such embodiments, the invention contemplates the alternating administration of a HSP preparation followed by the administration of a vaccine composition 4 to 6 days later, preferable 2 to 4 days, later, more preferably 1 to 2 days later, wherein such a cycle may be repeated as many times as desired. In certain embodiments, the heat shock protein preparation and vaccine composition are alternately administered in a cycle of less than 3 weeks, once every two weeks, once every 10 days or once every week.

The invention also contemplates the alternating administration of a o2M preparation followed by the administration of a vaccine composition 4 to 6 days later, preferable 2 to 4 days, later, more preferably 1 to 2 days later, wherein such a cycle may be repeated as many times as desired. In certain embodiments, the o2M preparation and vaccine composition are alternately administered in a cycle of less than 3 weeks, once every two weeks, once every 10 days or once every week.

In a specific embodiment, the HSP preparation or o2M preparation is administered to a subject within a time frame of one hour to twenty four hours after the administration of a vaccine. The time frame can be extended further to a few days or more if a slow-or continuous-release type of vaccine is used. This method is believed to help activate those APCs present in at or near the site of administration that may not yet have been activated by the presence of the vaccine.

In yet another specific embodiment, the HSP preparation or o2M preparation is administered to a subject within a time frame of about one to about twenty-four hours before the administration of a vaccine. In certain embodiments, the HSP preparation or o2M preparation is administered about 30 minutes to about 1 hour, about 1 to 2 hours, about 2 to 4 hours, about 4 to 6 hours, about 6 to 8 hours, about 8 to 10 hours, about 10 to 12 hours, about 12 to 14 hours, about 14 to 16 hours, about 16 to 20 hours, about 20 to 24 hours, about 24 to 36 hours, about 36 to 48 hours, up to about 56 hours before the administration of a vaccine. This method is believed to pre-activate the subject's APCs prior to the encounter with the vaccine.

In a preferred embodiment of the invention as provided above, the subject in whom the HSP preparation or o2M preparation and vaccine are administered is a human.

In yet another embodiment, the invention provides a method for inducing an immune response by a vaccine composition in a subject, wherein a sub-immunogenic amount of vaccine composition is used. As used herein, a sub-immunogenic amount of a vaccine composition refers to an amount that is insufficient for inducing an immune response if the vaccine composition is administered independent of the HSP preparation or o2M preparation. The method comprises administering to the subject an amount of a heat shock protein preparation or an amount of an o2M preparation before, concurrently with, or after the administration of the vaccine composition, such that said amount of vaccine composition effectively induces an immune response in the subject. In a preferred embodiment, the vaccine composition does not comprise a heat shock protein or an o2M.

In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the c2M preparation are not present in admixture.

In yet another embodiment, the invention provides a method of activating antigen presenting cells comprising contacting APCs with a heat shock protein preparation or an o2M preparation. Prior to treatment with a heat shock protein preparation or with an o2M preparation to activate the APCs, the cells can optionally be enriched or purified, and/or expanded ex vivo by methods well known in the art. The APCs can be obtained from a subject, preferably the same subject to whom the treated APCs are re-administered (i. e. , autologous APCs are used), although non-autologous APCs can also be used. The<BR> non-autologous APCs can be syngeneic (i. e. , from an identical twin of the individual to<BR> which the activated APCs will be administered) ; or allogeneic (i. e. , an individual who shares at least one common MHC allele with the individual to whom the activated APCs will be administered.) The activation of APCs can be monitored by techniques well known in the art, such as but not limited to those described in section 6 for testing CD1 lb+ cells. In the various embodiments as above-described, in the place of a HSP preparation or an o2M preparation, activated APCs can be administered to a subject for a similar result.

Accordingly, in a specific embodiment, the activated APCs can be used in vivo to produce or increase an immune response elicited by a vaccine composition which is administered to the subject at reasonably the same time. The activated APCs can alternatively be administered within a time frame of one to twenty four hours before or after the administration of a vaccine composition, or periodically for a few days or more-about 1 to 2 days, about 2 to 4 days, about 4 to 6 days, about 1 week, no more than 2 weeks after a slow-or continuous-release type of vaccine is used. Preferably, the treated APCs are administered to a site at or near the site of administration of the vaccine preparation. The administration of activated APCs can be conducted by any techniques known in the art.

In various embodiments of the invention, the HSP preparation may include but not limited to, hsp70, hsp90, gp96, singly or in combination with each other.

In other embodiments, each of the above embodiments may comprise administration of HSP preparation and o2M preparation in conjunction with a vaccine composition. In a preferred embodiment, the vaccine composition is not an HSP-peptide complex. In another preferred embodiment, the vaccine composition is not an o2M-peptide complex. In another preferred embodiment, the vaccine composition does not comprise a heat shock protein or an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition, the heat shock protein preparation and the o2M preparation are not present in admixture.

In various embodiments, the methods of the invention are used to treat or prevent any disease or disorder in which a therapeutic or prophylactic vaccine exists, i. e., that is amenable to treatment or prevention by an enhanced immune response. In specific embodiments the disease is an infectious disease, or a cancer. The heat shock protein preparation, a2M preparation or treated APCs are generally administered separately from the vaccine composition.

The invention includes methods for producing an immune response comprises administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced ; and administering to the subject a heat shock protein preparation, wherein the heat shock protein preparation does not elicit an immune response against the component in the absence of the administering of the vaccine composition. In a preferred embodiment, the vaccine composition does not

comprise a heat shock protein or an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the o2M preparation are not present in admixture.

The invention includes methods for producing an immune response comprises administering to the subject a vaccine composition comprising a component against which an immune response is desired to be induced; and administering to the subject a o2M preparation, wherein the o2M preparation does not elicit an immune response against the component in the absence of the administering of the vaccine composition. In a preferred embodiment, the vaccine composition does not comprise a heat shock protein or an o2M. In another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another preferred embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the o2M preparation are not present in admixture.

The invention encompasses methods for treating or preventing an infectious disease in a subject comprising in any order the steps of administering to the subject a vaccine composition comprising a component that displays the antigenicity of an infectious <BR> <BR> agent that causes the infectious disease (e. g. , an immunogenic amount of an antigen on the causative infectious agent); and administering to the subject an amount of a heat shock protein preparation effective in combination with the vaccine composition to induce or increase an immune response to the component in the subject, wherein the heat shock protein preparation does not elicit an immune response against said component in the absence of said administering of the vaccine composition. In a specific embodiment, the vaccine composition does not comprise a heat shock protein or an ou2M. In another specific embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another specific embodiment, if the component of the vaccine

composition is a peptide complexed to an od2M, the vaccine composition and the o2M preparation are not present in admixture.

The invention encompasses methods for treating or preventing an infectious disease in a subject comprising in any order the steps of administering to the subject a vaccine composition comprising a component that displays the antigenicity of an infectious <BR> <BR> agent that causes the infectious disease (e. g. , an immunogenic amount of an antigen on the causative infectious agent); and administering to the subject an amount of a o2M preparation effective in combination with the vaccine composition to induce or increase an immune response to the component in the subject, wherein the o2M preparation does not elicit an immune response against said component in the absence of said administering of the vaccine composition. In a specific embodiment, the vaccine composition does not comprise a heat shock protein or an o2M. In another specific embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another specific embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the o2M preparation are not present in admixture.

The invention also encompasses methods for treating or preventing a cancer or metastasis in a subject comprising in any order the steps of administering to the subject a vaccine composition comprising a component that displays the antigenicity of a cancer cell <BR> <BR> (e. g. , an immunogenic amount of an antigen on a cancer, such as but not limited to a tumor- specific antigen, and a tumor-associated antigen, or a molecule displaying antigenicity thereof) ; and administering to the subject an amount of a heat shock protein preparation effective to induce or increase an immune response in the subject to the component, wherein the heat shock protein preparation does not elicit an immune response against the component in the absence of the administering of the vaccine composition. In a specific embodiment, the vaccine composition does not comprise a heat shock protein or an o2M.

In another specific embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another specific embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the o2M preparation are not present in admixture.

The invention also encompasses methods for treating or preventing a cancer or metastasis in a subject comprising in any order the steps of administering to the subject a vaccine composition comprising a component that displays the antigenicity of a cancer cell (e. g. , an immunogenic amount of an antigen on a cancer, such as but not limited to a tumor- specific antigen, and a tumor-associated antigen, or a molecule displaying antigenicity thereof) ; and administering to the subject an amount of a o2M preparation effective to induce or increase an immune response in the subject to the component, wherein the o2M preparation does not elicit an immune response against the component in the absence of the administering of the vaccine composition. In a specific embodiment, the vaccine composition does not comprise a heat shock protein or an ol2M. In another specific embodiment, if the component of the vaccine composition is a peptide complexed to an heat shock protein, the vaccine composition and the heat shock protein preparation are not present in admixture. In yet another specific embodiment, if the component of the vaccine composition is a peptide complexed to an o2M, the vaccine composition and the o2M preparation are not present in admixture.

Another therapeutic method is also provided. In this embodiment, a mammalian (preferably human) HSP preparation or an c2M preparation is administered to a subject when it is desired that the APCs of the subject be in an activated state, such as when the subject is receiving a treatment modality that is not a vaccine. The HSP preparation or o2M preparation can be administered regularly for a period of time, e. g. , daily for up to several weeks, which may precede, overlap, and/or follow a treatment regimen with a non- vaccine modality. The HSP preparation or a2M preparation can be administered concurrently, before, or after the administration of the treatment modality. Examples of treatment modalities include but are not limited to antibiotics, antivirals, antifungal compounds, chemotherapeutic agents, and radiation, as well as biological therapeutic agents and immunotherapeutic agents. In one embodiment, the HSP preparation or o2M preparation can augment the therapeutic benefit of a treatment modality and improve the outcome of the treatment. Without being bound by any theory or mechanism, it is believed that the administration of a mammalian HSP preparation or o2M preparation to a subject can enhance the responsiveness of non-specific immune mechanisms of the subject, for example, by increasing the number of natural killer (NK) cells and/or accelerating the maturation of dendritic cells.

Where HSP-peptide complexes or o2M-peptide complexes are used, the peptides need not be antigenic or relevant to the condition in question. In this instance, the purpose of the invention is not to use a HSP-peptide complex or an o2M-peptide complex to elicit a specific immune response against the bound peptide. The HSP preparations and the o2M preparations of the invention generally aid presentation of all kinds of antigens in the subject, particularly those administered to the subject in the vaccine composition.

5.1 PREPARATION OF HEAT SHOCK PROTEINS Three major families of HSPs have been identified based on molecular weight. The families have been called hsp60, hsp70 and hsp90 where the numbers reflect the approximate molecular weight of the stress proteins in kilodaltons. Many members of these families were found subsequently to be induced in response to other stressful stimuli including, but not limited to, nutrient deprivation, metabolic disruption, oxygen radicals and infection with intracellular pathogens (See Welch, May 1993, Scientific American 56-64; Young, 1990, Annu. Rev. Immunol. 8: 401-420; Craig, 1993, Science 260: 1902-1903; <BR> <BR> Gething, et al. , 1992, Nature 355: 33-45; and Lindquist, et al. , 1988, Annu. Rev. Genetics 22: 631-677). A number of proteins thought to be involved in chaperoning functions are residents of the endoplasmic reticulum (ER) lumen and include, for example, protein <BR> <BR> disulfide isomerase (PDI; Gething et al. , 1992, Nature 355: 33-45), calreticulin (Herbert et<BR> al. , 1997, J. Cell Biol. 139: 613-623), Grp94 or ERp99 (Sorger & Pelham, 1987, J. Mol.

Biol. 194: (2) 341-4) which is related to hsp90, and Grp78 or BiP, which is related to hsp70 (Munro et al., 1986, Cell 46: 291-300; Haas & Webl, 1983, Nature 306: 387-389). It is contemplated that HSPs belonging to all of these three families, including fragments of such HSPs, can be used in the practice of the instant invention.

Heat shock proteins are among the most highly conserved proteins in existence. For example, DnaK, the hsp70 from E. coli has about 50% amino acid sequence identity with hsp70 proteins from excoriates (Bardwell, et al., 1984, Proc. Natl. Acad. Sci.

81: 848-852). The hsp60 and hsp90 families also show similarly high levels of intra families <BR> <BR> conservation (Hickey, et al. , 1989, Mol. Cell. Biol. 9: 2615-2626; Jindal, 1989, Mol. Cell.

Biol. 9: 2279-2283). In addition, it has been discovered that the hsp60, hsp70 and hsp90 families are composed of proteins that are related to the stress proteins in sequence, for example, having greater than 35% amino acid identity, but whose expression levels are not altered by stress. Therefore it is contemplated that the definition of stress protein, as used

herein, embraces other proteins, muteins, analogs, and variants thereof having at least 35% to 55%, preferably 55% to 75%, and most preferably 75% to 85% amino acid identity with members of the three families whose expression levels in a cell are enhanced in response to a stressful stimulus. The purification of stress proteins belonging to these three families is described below.

In addition, HSPs have been found to have immunological and antigenic properties. HSPs are now understood to play an essential role in immune regulation. For instance, prior experiments have demonstrated that HSPs stimulate strong and long-lasting specific immune responses against antigenic peptides that have been covalently or noncovalently attached to the HSPs. By utilizing a specific peptide, the immune response generated is"specific"or targeted to that peptide.

In the present invention, purified unbound HSPs, HSPs covalently or noncovalently bound to specific peptides or nonspecific peptides (collectively referred to herein as HSP-peptide complexes), and combinations of thereof are used. Purification of HSPs in complexed or non-complexed forms are described in the following subsections.

Further, one skilled in the art can synthesize HSPs by recombinant expression or peptide synthesis, which are also described below. In the present invention, an HSP preparation can comprise unbound hsp70, hsp90, gp96, calreticulin, hsplO0 or grpl70 or noncovalent or covalent complexes thereof complexed to a peptide.

5.1. 1 Preparation and Purification of Hsp70 or Hsp70-peptide Complexes The purification of hsp70-peptide complexes has been described previously, see, for example, Udono et al. , 1993, J. Exp. Med. 178: 1391-1396. A procedure that may be used, presented by way of example but not limitation, is as follows: Initially, human or mammalian cells are suspended in 3 volumes of 1X Lysis buffer consisting of 5mM sodium phosphate buffer (pH 7), 150mM NaCI, 2mM CaCl2, 2mM MgCl2 and 1mM phenyl methyl sulfonyl fluoride (PMSF). Then, the pellet is sonicated, on ice, until >99% cells are lysed as determined by microscopic examination. As an alternative to sonication, the cells may be lysed by mechanical shearing and in this approach the cells typically are resuspended in 30mM sodium bicarbonate (pH 7.5), ImM PMSF, incubated on ice for 20 minutes and then homogenized in a Dounce homogenizer until >95% cells are lysed.

Then the lysate is centrifuged at 1, 000g for 10 minutes to remove unbroken cells, nuclei and other cellular debris. The resulting supernatant is recentrifuged at 100,000g for 90 minutes, the supernatant harvested and then mixed with Con A Sepharose equilibrated with phosphate buffered saline (PBS) containing 2mM Ca2+ and 2mM Mg2+. When the cells are lysed by mechanical shearing the supernatant is diluted with an equal volume of 2X lysis buffer prior to mixing with Con A Sepharose. The supernatant is then allowed to bind to the Con A Sepharose for 2-3 hours at 4°C. The material that fails to bind is harvested and dialyzed for 36 hours (three times, 100 volumes each time) against lOmM Tris-Acetate (pH 7.5), 0. 1mM EDTA, lOmM NaCI, 1mM PMSF.

Then the dialyzate is centrifuged at 17,000 rpm (Sorvall SS34 rotor) for 20 minutes. Then the resulting supernatant is harvested and applied to a Mono Q FPLCTM ion exchange chromatographic column (Pharmacia) equilibrated in 20mM Tris-Acetate (pH 7.5), 20mM NaCI, O. 1mM EDTA and 15mM 2-mercaptoethanol. The column is then developed with a 20mM to 500mM NaCI gradient and then eluted fractions fractionated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and characterized by immunoblotting using an appropriate anti-hsp70 antibody (such as from clone N27F3-4, from StressGen).

Fractions strongly immunoreactive with the anti-hsp70 antibody are pooled and the hsp70-peptide complexes precipitated with ammonium sulfate; specifically with a 50%-70% ammonium sulfate cut. The resulting precipitate is then harvested by centrifugation at 17,000 rpm (SS34 Sorvall rotor) and washed with 70% ammonium sulfate.

The washed precipitate is then solubilized and any residual ammonium sulfate removed by gel filtration on a Sephadex G25 column (Pharmacia). If necessary the hsp70 preparation thus obtained can be repurified through the Mono Q FPLCTM ion exchange chromatographic column (Pharmacia) as described above.

The hsp70-peptide complex can be purified to apparent homogeneity using this method. Typically 1 mg of hsp70-peptide complex can be purified from 1 g of cells/tissue.

An improved method for purification of hsp70-peptide complexes comprises contacting cellular proteins with ADP or a nonhydrolyzable analog of ATP affixed to a solid substrate, such that hsp70 in the lysate can bind to the ADP or nonhydrolyzable ATP analog, and eluting the bound hsp70. A preferred method uses column chromatography

with ADP affixed to a solid substratum (e. g., ADP-agarose). The resulting hsp70 preparations are higher in purity and devoid of contaminating peptides. The hsp70 complex yields are also increased significantly by about more than 10 fold. Alternatively, chromatography with nonhydrolyzable analogs of ATP, instead of ADP, can be used for purification of hsp70-peptide complexes. By way of example but not limitation, purification of hsp70-peptide complexes by ADP-agarose chromatography can be carried out as follows: Meth A sarcoma cells (500 million cells) are homogenized in hypotonic buffer and the lysate is centrifuged at 100,000 g for 90 minutes at 4°C. The supernatant is applied to an ADP-agarose column. The column is washed in buffer and is eluted with 5 column volumes of 3 mM ADP. The hsp70-peptide complexes elute in fractions 2 through 10 of the total 15 fractions which elute. The eluted fractions are analyzed by SDS-PAGE.

The hsp70-peptide complexes can be purified to apparent homogeneity using this procedure.

Separation of the HSP from an hsp70-peptide complex can be performed in the presence of ATP or low pH. These two methods may be used to elute the peptide from an hsp70-peptide complex. The first approach involves incubating an hsp70-peptide complex preparation in the presence of ATP. The other approach involves incubating an hsp70-peptide complex preparation in a low pH buffer. These methods and any others known in the art may be applied to separate the HSP and peptide from an hsp-peptide complex.

5.1. 2 Preparation and Purification of Hsp90 or Hsp90-peptide Complexes A procedure that can be used, presented by way of example and not limitation, is as follows: Initially, human or mammalian cells are suspended in 3 volumes of 1X Lysis buffer consisting of 5mM sodium phosphate buffer (pH 7), 150mM NaCI, 2mM CaCl2, 2mM MgCl2 and 1mM phenyl methyl sulfonyl fluoride (PMSF). Then, the pellet is sonicated, on ice, until >99% cells are lysed as determined by microscopic examination. As an alternative to sonication, the cells may be lysed by mechanical shearing and in this approach the cells typically are resuspended in 30mM sodium bicarbonate (pH 7.5), 1mM

PMSF, incubated on ice for 20 minutes and then homogenized in a Dounce homogenizer until >95% cells are lysed.

Then the lysate is centrifuged at 1, 000g for 10 minutes to remove unbroken cells, nuclei and other cellular debris. The resulting supernatant is recentrifuged at 100,000g for 90 minutes, the supernatant harvested and then mixed with Con A Sepharose equilibrated with PBS containing 2mM Ca2+ and 2mM Mg2+. When the cells are lysed by mechanical shearing the supernatant is diluted with an equal volume of 2X Lysis buffer prior to mixing with Con A Sepharose. The supernatant is then allowed to bind to the Con A Sepharose for 2-3 hours at 4°C. The material that fails to bind is harvested and dialyzed for 36 hours (three times, 100 volumes each time) against lOmM Tris-Acetate (pH 7.5), 0. 1mM EDTA, lOmM NaCI, ImM PMSF. Then the dialyzate is centrifuged at 17,000 rpm (Sorvall SS34 rotor) for 20 minutes. Then the resulting supernatant is harvested and applied to a Mono Q FPLCTM ion exchange chromatographic column (Pharmacia) equilibrated with lysis buffer. The proteins are then eluted with a salt gradient of 200mM to 600mM NaCI.

The eluted fractions are fractionated by SDS-PAGE and fractions containing the hsp90-peptide complexes identified by immunoblotting using an anti-hsp90 antibody such as 3G3 (Affinity Bioreagents). Hsp90-peptide complexes can be purified to apparent homogeneity using this procedure. Typically, 150-200, ug of hsp90-peptide complex can be purified from I g of cells/tissue.

Separation of the HSP from an hsp90-peptide complex can be performed in the presence of ATP or low pH. These two methods may be used to elute the peptide from an hsp90-peptide complex. The first approach involves incubating an hsp90-peptide complex preparation in the presence of ATP. The other approach involves incubating an hsp90-peptide complex preparation in a low pH buffer. These methods and any others known in the art may be applied to separate the HSP and peptide from an hsp-peptide complex.

5.1. 3 Preparation and Purification of Gp96 or Gp96-peptide Complexes A procedure that can be used, presented by way of example and not limitation, is as follows:

A pellet of human or mammalian cells is resuspended in 3 volumes of buffer consisting of 30mM sodium bicarbonate buffer (pH 7.5) and 1mM PMSF and the cells allowed to swell on ice 20 minutes. The cell pellet is then homogenized in a Dounce homogenizer (the appropriate clearance of the homogenizer will vary according to each cell type) on ice until >95% cells are lysed.

The lysate is centrifuged at 1, OOOg for 10 minutes to remove unbroken cells, nuclei and other debris. The supernatant from this centrifugation step is then recentrifuged at 100,000g for 90 minutes. The gp96-peptide complex can be purified either from the 100,000 pellet or from the supernatant.

When purified from the supernatant, the supernatant is diluted with equal volume of 2X lysis buffer and the supernatant mixed for 2-3 hours at 4°C with Con A Sepharose equilibrated with PBS containing 2mM Ca2+ and 2mM Mg2+. Then, the slurry is packed into a column and washed with 1X lysis buffer until the OD2go drops to baseline.

Then, the column is washed with 1/3 column bed volume of 10% a-methyl mannoside (a MM) dissolved in PBS containing 2mM Ca2+ and 2mM Mg2+, the column sealed with a piece of parafilm, and incubated at 37°C for 15 minutes. Then the column is cooled to room temperature and the parafilin removed from the bottom of the column. Five column volumes of the a-MM buffer are applied to the column and the eluate analyzed by SDS- PAGE. Typically the resulting material is about 60-95% pure, however this depends upon the cell type and the tissue-to-lysis buffer ratio used. Then the sample is applied to a Mono Q FPLCTM ion exchange chromatographic column (Pharmacia) equilibrated with a buffer containing 5mM sodium phosphate (pH 7). The proteins are then eluted from the column with a 0-1M NaCI gradient and the gp96 fraction elutes between 400mM and 550mM NaCl.

The procedure, however, may be modified by two additional steps, used either alone or in combination, to consistently produce apparently homogeneous gp96- peptide complexes. One optional step involves an ammonium sulfate precipitation prior to the Con A purification step and the other optional step involves DEAE-Sepharose purification after the Con A purification step but before the Mono Q FPLCTM step.

In the first optional step, described by way of example as follows, the supernatant resulting from the 100,000g centrifugation step is brought to a final concentration of 50% ammonium sulfate by the addition of ammonium sulfate. The ammonium sulfate is added slowly while gently stirring the solution in a beaker placed in a

tray of ice water. The solution is stirred from about % 2 to 12 hours at 4°C and the resulting solution centrifuged at 6,000 rpm (Sorvall SS34 rotor). The supernatant resulting from this step is removed, brought to 70% ammonium sulfate saturation by the addition of ammonium sulfate solution, and centrifuged at 6,000 rpm (Sorvall SS34 rotor). The resulting pellet from this step is harvested and suspended in PBS containing 70% ammonium sulfate in order to rinse the pellet. This mixture is centrifuged at 6,000 rpm (Sorvall SS34 rotor) and the pellet dissolved in PBS containing 2mM Ca2+ and Mg2+.

Undissolved material is removed by a brief centrifugation at 15,000 rpm (Sorvall SS34 rotor). Then, the solution is mixed with Con A Sepharose and the procedure followed as before.

In the second optional step, described by way of example as follows, the gp96 containing fractions eluted from the Con A column are pooled and the buffer exchanged for 5mM sodium phosphate buffer (pH 7), 300mM NaCl by dialysis, or preferably by buffer exchange on a Sephadex G25 column. After buffer exchange, the solution is mixed with DEAE-Sepharose previously equilibrated with 5mM sodium phosphate buffer (pH 7), 300mM NaCl. The protein solution and the beads are mixed gently for 1 hour and poured into a column. Then, the column is washed with 5mM sodium phosphate buffer (pH 7), 300mM NaCl, until the absorbance at 280nm drops to baseline.

Then, the bound protein is eluted from the column with five volumes of 5mM sodium phosphate buffer (pH 7), 700mM NaCl. Protein containing fractions are pooled and diluted with 5mM sodium phosphate buffer (pH 7) in order to lower the salt concentration to 175mM. The resulting material then is applied to the Mono Q FPLCTM ion exchange chromatographic column (Pharmacia) equilibrated with 5mM sodium phosphate buffer (pH 7) and the protein that binds to the Mono Q FPLCTM ion exchange chromatographic column (Pharmacia) is eluted as described before.

It is appreciated, however, that one skilled in the art may assess, by routine experimentation, the benefit of incorporating the second optional step into the purification protocol. In addition, it is appreciated also that the benefit of adding each of the optional steps will depend upon the source of the starting material.

When the gp96 fraction is isolated from the 100,000g pellet, the pellet is suspended in 5 volumes of PBS containing either 1% sodium deoxycholate or 1% oxtyl glucopyranoside (but without the Mg2+ and Ca2+) and incubated on ice for 1 hour. The

suspension is centrifuged at 20, 000g for 30 minutes and the resulting supernatant dialyzed against several changes of PBS (also without the Mg2+ and Ca2+) to remove the detergent.

The dialysate is centrifuged at 100,000g for 90 minutes, the supernatant harvested, and calcium and magnesium are added to the supernatant to give final concentrations of 2mM, respectively. Then the sample is purified by either the unmodified or the modified method for isolating gp96-peptide complex from the 100,000g supernatant, see above.

The gp96-peptide complexes can be purified to apparent homogeneity using this procedure. About 10-20yg of gp96 can be isolated from lg cells/tissue.

Separation of the HSP from an gp96-peptide complex can be performed in the presence of ATP or low pH. These two methods may be used to elute the peptide from an gp96-peptide complex. The first approach involves incubating an gp96-peptide complex preparation in the presence of ATP. The other approach involves incubating an gp96- peptide complex preparation in a low pH buffer. These methods and any others known in the art may be applied to separate the HSP and peptide from an hsp-peptide complex.

5.1. 4 Preparation and Purification of Hspl 10-peptide Complexes A procedure, described by Wang et al., 2001, J. Immunol. 166 (1) : 490-7, that can be used, presented by way of example and not limitation, is as follows: A pellet (40-60 ml) of cell or tissue, e. g., tumor cell tissue, is homogenized in 5 vol of hypotonic buffer (30 mN sodium bicarbonate, pH7.2, and protease inhibitors) by Dounce homogenization. The lysate is centrifuged at 4,500 x g and then 100,000 x g for 2 hours. If the cells or tissues are of hepatic origin, the resulting supernatant is was first applied to a blue Sepharose column (Pharmacia) to remove albumin. Otherwise, the resulting supernatant is applied to a Con A-Sepharose column (Pharmacia Biotech, Piscataway, NJ) previously equilibrated with binding buffer (20mM Tris-HCI, pH 7.5 ; 100mM NaCl ; lmM MgCk ; 1 mM CaCl2 ; 1 mM MnCl2 ; and 15 mM 2-ME). The bound proteins are eluted with binding buffer containing 15% a-D-o-methylmannoside (Sigma, St.

Louis, MO).

Con A-Sepharose unbound material is first dialyzed against a solution of 20 mM Tris-HCI, pH 7.5 ; 100 mM NaCI ; and 15 mM 2-ME, and then applied to a DEAE- Sepharose column and eluted by salt gradient from 100 to 500 mM NaCI. Fractions containing hspl 10 are collected, dialyzed, and loaded onto a Mono Q (Pharmacia) 10/10

column equilibrated with 20mM Tris-HCl, pH 7.5 ; 200 mM NaCI ; and 15 mM 2-ME. The bound proteins are eluted with a 200-500 mM NaCI gradient. Fractions are analyzed by SDS-PAGE followed by immunoblotting with an Ab for hspl 10, as described by Wang et al., 1999, J. Immunol. 162: 3378. Pooled fractions containing hspl 10 are concentrated by Centriplus (Amicon, Beverly, MA) and applied to a Superose 12 column (Pharmacia).

Proteins are eluted by 40 mM Tris-HCl, pH 8. 0; 150 mM NaCl ; and 15 mM 2-ME with a flow rate of 0.2 ml/min.

5.1. 5 Preparation and Purification of ProducedGrp 70-peptide Complexes A procedure, described by Wang et al., 2001, J. Immunol. 166 (1) : 490-7, that can be used, presented by way of example and not limitation, is as follows: A pellet (40-60 ml) of cell or tissue, e. g., tumor cell tissue, is homogenized in 5 vol of hypotonic buffer (30 mN sodium bicarbonate, pH7.2, and protease inhibitors) by Dounce homogenization. The lysate is centrifuged at 4,500 x g and then 100,000 x g for 2 hours. If the cells or tissues are of hepatic origin, the resulting supernatant is was first applied to a blue Sepharose column (Pharmacia) to remove albumin. Otherwise, the resulting supernatant is applied to a Con A-Sepharose column (Pharmacia Biotech, Piscataway, NJ) previously equilibrated with binding buffer (20mM Tris-HCI, pH 7.5 ; 100mM NaCl ; 1mM MgCk ; 1 mM CaCl2 ; 1 mM MnCl2 ; and 15 mM 2-ME). The bound proteins are eluted with binding buffer containing 15% a-D-o-methylmannoside (Sigma, St.

Louis, MO).

Con A-Sepharose-bound material is first dialyzed against 20 mM Tris-HCl, pH 7.5, and 150 mM NaCI and then applied to a Mono Q column and eluted by a 150 to 400 mM NaCI gradient. Pooled fractions are concentrated and applied on the Superose 12 column (Pharmacia). Fractions containing homogeneous grip 170 are collected.

5.1. 6 Recombinant Expression of HSPs Methods known in the art can be utilized to recombinantly produce HSPs. A nucleic acid sequence encoding a heat shock protein can be inserted into an expression vector for propagation and expression in host cells.

An expression construct, as used herein, refers to a nucleotide sequence encoding an HSP operably associated with one or more regulatory regions which enables expression of the HSP in an appropriate host cell. "Operably-associated"refers to an

association in which the regulatory regions and the HSP sequence to be expressed are joined and positioned in such a way as to permit transcription, and ultimately, translation.

The regulatory regions necessary for transcription of the HSP can be provided by the expression vector. A translation initiation codon (ATG) may also be provided if the HSP gene sequence lacking its cognate initiation codon is to be expressed.

In a compatible host-construct system, cellular transcriptional factors, such as RNA polymerase, will bind to the regulatory regions on the expression construct to effect transcription of the modified HSP sequence in the host organism. The precise nature of the regulatory regions needed for gene expression may vary from host cell to host cell.

Generally, a promoter is required which is capable of binding RNA polymerase and promoting the transcription of an operably-associated nucleic acid sequence. Such regulatory regions may include those 5'non-coding sequences involved with initiation of transcription and translation, such as the TATA box, capping sequence, CAAT sequence, and the like. The non-coding region 3'to the coding sequence may contain transcriptional termination regulatory sequences, such as terminators and polyadenylation sites.

In order to attach DNA sequences with regulatory functions, such as promoters, to the HSP gene sequence or to insert the HSP gene sequence into the cloning site of a vector, linkers or adapters providing the appropriate compatible restriction sites may be ligated to the ends of the cDNAs by techniques well known in the art (Wu et al., 1987, Methods in Enzymol 152: 343-349). Cleavage with a restriction enzyme can be followed by modification to create blunt ends by digesting back or filling in single-stranded DNA termini before ligation. Alternatively, a desired restriction enzyme site can be introduced into a fragment of DNA by amplification of the DNA by use of PCR with primers containing the desired restriction enzyme site.

An expression construct comprising an HSP sequence operably associated with regulatory regions can be directly introduced into appropriate host cells for expression and production of HSP-peptide complexes without further cloning. See, for example, U. S.

Patent No. 5,580, 859. The expression constructs can also contain DNA sequences that facilitate integration of the HSP sequence into the genome of the host cell, e. g. , via homologous recombination. In this instance, it is not necessary to employ an expression vector comprising a replication origin suitable for appropriate host cells in order to propagate and express the HSP in the host cells.

A variety of expression vectors may be used including, but not limited to, plasmids, cosmids, phage, phagemids or modified viruses. Typically, such expression vectors comprise a functional origin of replication for propagation of the vector in an appropriate host cell, one or more restriction endonuclease sites for insertion of the HSP gene sequence, and one or more selection markers. The expression vector must be used with a compatible host cell which may be derived from a prokaryotic or an eukaryotic organism including but not limited to bacteria, yeasts, insects, mammals and humans.

For long term, high yield production of properly processed HSP or HSP- peptide complexes, stable expression in mammalian cells is preferred. Cell lines that stably express HSP or HSP-peptide complexes may be engineered by using a vector that contains a selectable marker. By way of example but not limitation, following the introduction of the expression constructs, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the expression construct confers resistance to the selection and optimally allows cells to stably integrate the expression construct into their chromosomes and to grow in culture and to be expanded into cell lines. Such cells can be cultured for a long period of time while HSP is expressed continuously.

The recombinant cells may be cultured under standard conditions of temperature, incubation time, optical density and media composition. However, conditions for growth of recombinant cells may be different from those for expression of HSPs and antigenic proteins. Modified culture conditions and media may also be used to enhance production of the HSP. For example, recombinant cells containing HSPs with their cognate promoters may be exposed to heat or other environmental stress, or chemical stress. Any techniques known in the art may be applied to establish the optimal conditions for producing HSP or HSP-peptide complexes.

5.1. 7 Peptide Synthesis An alternative to producing HSP by recombinant techniques is peptide synthesis. For example, an entire HSP, or a peptide corresponding to a portion of an HSP can be synthesized by use of a peptide synthesizer. Conventional peptide synthesis or other synthetic protocols well known in the art may be used.

Peptides having the amino acid sequence of a HSP or a portion thereof may be synthesized by solid-phase peptide synthesis using procedures similar to those described

by Merrifield, 1963, J. Am. Chem. Soc. , 85: 2149. During synthesis, N-a-protected amino acids having protected side chains are added stepwise to a growing polypeptide chain linked by its C-terminal and to an insoluble polymeric support i. e. , polystyrene beads. The peptides are synthesized by linking an amino group of an N-a-deprotected amino acid to an a-carboxyl group of an N-a-protected amino acid that has been activated by reacting it with a reagent such as dicyclohexylcarbodiimide. The attachment of a free amino group to the activated carboxyl leads to peptide bond formation. The most commonly used N-a- protecting groups include Boc which is acid labile and Fmoc which is base labile. Details of appropriate chemistries, resins, protecting groups, protected amino acids and reagents are well known in the art and so are not discussed in detail herein (See, Atherton, et al., 1989, Solid Phase Peptide Synthesis: A Practical Approach, IRL Press, and Bodanszky, 1993, Peptide Chemistry, A Practical Textbook, 2nd Ed. , Springer-Verlag).

Purification of the resulting HSP is accomplished using conventional procedures, such as preparative HPLC using gel permeation, partition and/or ion exchange chromatography. The choice of appropriate matrices and buffers are well known in the art and so are not described in detail herein.

5.2 o2-MACROGLOBULIN Alpha-2-macroglobulin can be bought from commercial sources or prepared by purifying it from human blood. To purify o2M from blood, the following non-limiting protocol can be used: Blood is collected from a subject and is allowed to clot. It is then centrifuged for 30 minutes under 14,000 x g to obtain the serum which is then applied to a gel filtration column (Sephacryl S-300R) equilibrated with 0.04M Tris buffer pH 7.6 plus 0.3M NaCl. A 65ml column is used for about 10ml of serum. Three ml fractions are collected and each fraction is tested for the presence of o2M by dot blot using an o2M specific antibody. The o2M positive fractions are pooled and applied to a PD10 column to exchange the buffer to. 01M Sodium Phosphate buffer pH 7.5 with PMSF. The pooled fractions are then applied to a Con A column (lOml) equilbrated with the phosphate buffer.

The column is washed and the protein is eluted with 5% methylmannose pyranoside. The eluent is passed over a PD10 column to change the buffer to a Sodium Acetate buffer (0. 05M ; pH6.0). A DEAE column is then equilibrated with acetate buffer and the sample is

applied to the DEAE column. The column is washed and the protein is eluted with 0.13M sodium acetate. The fractions with o2M are then pooled.

As an alternative to using HSP/o2M and the peptides with which they are associated, the following methods can be followed to produce complexes in vitro.

5.2. 1 Recombinant Expression of Heat Shock Proteins and o2M In certain embodiments of the present invention, HSPs and o2M can be prepared from cells that express higher levels of HSPs and ot2M through recombinant means. Amino acid sequences and nucleotide sequences of many HSPs and o2M are generally available in sequence databases, such as GenBank. Computer programs, such as Entrez, can be used to browse the database, and retrieve any amino acid sequence and genetic sequence data of interest by accession number. These databases can also be searched to identify sequences with various degrees of similarities to a query sequence using programs, such as FASTA and BLAST, which rank the similar sequences by alignment scores and statistics. Such nucleotide sequences of non-limiting examples of HSPs that can be used for the compositions, methods, and for preparation of the HSP peptide-complexs of the invention are as follows: human HSP70, Genbank Accession No. M24743, Hunt et al., 1995, Proc. Natl. Acad. Sci. U. S. A. , 82: 6455-6489 ; human HSP90, Genbank Accession No. X15183, Yamazaki et al., Nucl. Acids Res. 17: 7108; human gp96: Genbank Accession No. X15187, Maki et al., 1990, Proc. Natl. Acad. Sci.

U. S. A. 87 : 5658-5562; human BiP: Genbank Accession No. M19645; Ting et al., 1988, DNA 7 : 275-286 ; human HSP27, Genbank Accession No. M24743; Hickey et al., 1986, Nucleic Acids Res. 14: 4127-45; mouse HSP70: Genbank Accession No. M35021, Hunt et al., 1990, Gene 87: 199-204; mouse gp96: Genbank Accession No. M16370, Srivastava et al., 1987, Proc. Natl. Acad. Sci. U. S. A. 85: 3807-3811 ; and mouse BiP: Genbank Accession No. U16277, Haas et al., 1988, Proc. Natl. Acad. Sci. U. S. A. 85 : 2250-2254. Due to the degeneracy of the genetic code, the term"HSP gene", as used herein, refers not only to the naturally occurring nucleotide sequence but also encompasses all the other degenerate DNA sequences that encode the HSP.

As used herein, the term"a2M"embraces other polypeptide fragments, analogs, and variants of c2M having at least 35% to 55%, preferably 55% to 75%, and most preferably 75% to 85% amino acid identity with c2M, and is capable of forming a complex with an antigenic peptide, which complex is capable of being taken up by an antigen

presenting cell and eliciting an immune response against the antigenic molecule. The o2M molecule of the invention can be purchased commercially or purified from natural sources (Kurecki et al., 1979, Anal. Biochem. 99: 415-420), chemically synthesized, or recombinantly produced. Non-limiting examples of o2M sequences that can be used for preparation of the o2M polypeptides of the invention are as follows: Genbank Accession Nos. M11313, P01023, AAA51551 ; Kanetal., 1985, Proc. Nat. Acad. Sci. 82: 2282-2286. Due to the degeneracy of the genetic code, the term"o2M gene", as used herein, refers not only to the naturally occurring nucleotide sequence but also encompasses all the other degenerate DNA sequences that encode an o2M polypeptide.

Once the nucleotide sequence encoding the HSP or o2M of choice has been identified, the nucleotide sequence, or a fragment thereof, can be obtained and cloned into an expression vector for recombinant expression. The expression vector can then be introduced into a host cell for propogation of the HSP or o2M. Methods for recombinant production of HSPs or c2M are described in detail herein.

The DNA may be obtained by DNA amplification or molecular cloning <BR> <BR> directly from a tissue, cell culture, or cloned DNA (e. g. , a DNA"library") using standard molecular biology techniques (see e. g., Methods in Enzymology, 1987, volume 154, Academic Press; Sambrook et al. 1989, Molecular Cloning-A Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, New York; and Current Protocols in Molecular Biology, <BR> <BR> Ausubel et al. (eds. ), Greene Publishing Associates and Wiley Interscience, New York, each of which is incorporated herein by reference in its entirety). Clones derived from genomic DNA may contain regulatory and intron DNA regions in addition to coding regions; clones derived from cDNA will contain only exon sequences. Whatever the source, the HSP or o2M gene should be cloned into a suitable vector for propagation of the gene.

In a preferred embodiment, DNA can be amplified from genomic or cDNA by polymerase chain reaction (PCR) amplification using primers designed from the known sequence of a related or homologous HSP or ol2M. PCR is used to amplify the desired sequence in DNA clone or a genomic or cDNA library, prior to selection. PCR can be carried out, e. g. , by use of a thermal cycler and Taq polymerase (Gene Amp@). The polymerase chain reaction (PCR) is commonly used for obtaining genes or gene fragments of interest. For example, a nucleotide sequence encoding an HSP or o2M of any desired length can be generated using PCR primers that flank the nucleotide sequence encoding

open reading tram. Alternatively, an HSP or o2M gene sequence can be cleaved at appropriate sites with restriction endonuclease (s) if such sites are available, releasing a fragment of DNA encoding the HSP or o2M gene. If convenient restriction sites are not available, they may be created in the appropriate positions by site-directed mutagenesis and/or DNA amplification methods known in the art (see, for example, Shankarappa et al., 1992, PCR Method Appl. 1: 277-278). The DNA fragment that encodes the HSP or c2M is then isolated, and ligated into an appropriate expression vector, care being taken to ensure that the proper translation reading frame is maintained.

In an alternative embodiment, for the molecular cloning of an HSP or o2M gene from genomic DNA, DNA fragments are generated to form a genomic library. Since some of the sequences encoding related HSPs or a2M are available and can be purified and labeled, the cloned DNA fragments in the genomic DNA library may be screened by nucleic acid hybridization to a labeled probe (Benton and Davis, 1977, Science 196: 180; Grunstein and Hogness, 1975, Proc. Natl. Acad. Sci. U. S. A. 72: 3961). Those DNA fragments with substantial homology to the probe will hybridize. It is also possible to identify an appropriate fragment by restriction enzyme digestion (s) and comparison of fragment sizes with those expected according to a known restriction map.

Alternatives to isolating the HSP or o2M genomic DNA include, but are not limited to, chemically synthesizing the gene sequence itself from a known sequence or synthesizing a cDNA to the mRNA which encodes the HSP or c2M. For example, RNA for cDNA cloning of the HSP or o2M gene can be isolated from cells which express the HSP or o2M. A cDNA library may be generated by methods known in the art and screened by methods, such as those disclosed for screening a genomic DNA library. If an antibody to the HSP or o2M is available, the HSP or o2M may be identified by binding of a labeled antibody to the HSP-or oc2M-synthesizing clones.

Other specific embodiments for the cloning of a nucleotide sequence encoding an HSP or o2M, are presented as examples but not by way of limitation, as follows: In a specific embodiment, nucleotide sequences encoding an HSP or o2M can be identified and obtained by hybridization with a probe comprising a nucleotide sequence encoding HSP or o2M under conditions of low to medium stringency. By way of example and not limitation, procedures using such conditions of low stringency are as follows (see also Shilo and Weinberg, 1981, Proc. Natl. Acad. Sci. U. S. A. 78: 6789-6792). Filters

ELISA and FACS analysis and its purification by affinity chromatography with the aid of Protein A.

5.2. 1.1 Expression Systems Nucleotide sequences encoding an HSP or o2M molecule can be inserted into the expression vector for propagation and expression in recombinant cells. An expression construct, as used herein, refers to a nucleotide sequence encoding an HSP or a2M operably associated with one or more regulatory regions which allows expression of the HSP or o2M molecule in an appropriate host cell. "Operably-associated"refers to an association in which the regulatory regions and the HSP or c2M polypeptide sequence to be expressed are joined and positioned in such a way as to permit transcription, and ultimately, translation of the HSP or o2M sequence. A variety of expression vectors may be used for the expression of HSPs or c2M, including, but not limited to, plasmids, cosmids, phage, phagemids, or modified viruses. Examples include bacteriophages such as lambda derivatives, or plasmids such as pBR322 or pUC plasmid derivatives or the Bluescript vector (Stratagene). Typically, such expression vectors comprise a functional origin of replication for propagation of the vector in an appropriate host cell, one or more restriction endonuclease sites for insertion of the HSP or o2M gene sequence, and one or more selection markers.

For expression of HSPs or o2M in mammalian host cells, a variety of regulatory regions can be used, for example, the SV40 early and late promoters, the cytomegalovirus (CMV) immediate early promoter, and the Rous sarcoma virus long terminal repeat (RSV-LTR) promoter. Inducible promoters that may be useful in mammalian cells include but are not limited to those associated with the metallothionein II gene, mouse mammary tumor virus glucocorticoid responsive long terminal repeats (MMTV-LTR), the 0-interferon gene, and the HSP70 gene (Williams et al., 1989, Cancer Res. 49: 2735-42; Taylor et al., 1990, Mol. Cell. Biol. 10: 165-75). The efficiency of expression of the HSP or o2M in a host cell may be enhanced by the inclusion of appropriate transcription enhancer elements in the expression vector, such as those found in SV40 virus, Hepatitis B virus, cytomegalovirus, immunoglobulin genes, metallothionein, 0- actin (see Bittner et al., 1987, Methods in Enzymol. 153: 516-544; German, 1990, Curr. Op. in Biotechnol. 1 : 36-47).

The expression vector may also contain sequences that permit maintenance and replication of the vector in more than one type of host cell, or integration of the vector into the host chromosome. Such sequences may include but are not limited to replication origins, autonomously replicating sequences (ARS), centromere DNA, and telomere DNA.

It may also be advantageous to use shuttle vectors that can be replicated and maintained in at least two types of host cells.

In addition, the expression vector may contain selectable or screenable marker genes for initially isolating or identifying host cells that contain DNA encoding an HSP or o2M. For long term, high yield production of HSPs or o2M, stable expression in mammalian cells is preferred. A number of selection systems may be used for mammalian cells, including, but not limited, to the Herpes simplex virus thymidine kinase (Wigler et al., 1977, Cell 11: 223), hypoxanthine-guanine phosphoribosyltransferase (Szybalski and Szybalski, 1962, Proc. Natl. Acad. Sci. U. S. A. 48: 2026), and adenine phosphoribosyltransferase (Lowy et al., 1980, Cell 22: 817) genes can be employed in tk, hgprt or aprf cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for dihydrofolate reductase (dhfr), which confers resistance to methotrexate (Wigler et al., 1980, Natl. Acad. Sci. U. S. A. 77 : 3567 ; O'Hare et al., 1981, Proc. Natl.

Acad. Sci. U. S. A. 78 : 1527); gpt, which confers resistance to mycophenolic acid (Mulligan and Berg, 1981, Proc. Natl. Acad. Sci. U. S. A. 78 : 2072); neomycin phosphotransferase (neo), which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., 1981, J. Mol. Biol. 150: 1); and hygromycin phosphotransferase (hyg), which confers resistance to hygromycin (Santerre et al., 1984, Gene 30: 147). Other selectable markers, such as but not limited to histidinol and Zeocin can also be used.

The expression construct comprising an HSP-or c2M-coding sequence operably associated with regulatory regions can be directly introduced into appropriate host cells for expression and production of the HSP or o2M complexes of the invention without further cloning (see, for example, U. S. Patent No. 5, 580, 859). The expression constructs may also contain DNA sequences that facilitate integration of the coding sequence into the genome of the host cell, e. g., via homologous recombination. In this instance, it is not necessary to employ an expression vector comprising a replication origin suitable for appropriate host cells in order to propagate and express the HSP or o2M molecule in the host cells.

containing DNA are pretreated for 6 h at 40°C in a solution containing 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0. 1% PVP, 0. 1% Ficoll, 1% BSA, and 500 , ug/ml denatured salmon sperm DNA. Hybridizations are carried out in the same solution with the following modifications: 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100, ug/ml salmon sperm DNA, 10% (wt/vol) dextran sulfate, and 5-20 X 106 cpm 32P-labeled probe is used.

Filters are incubated in hybridization mixture for 18-20 h at 40°C, and then washed for 1.5 h at 55°C in a solution containing 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0. 1% SDS. The wash solution is replaced with fresh solution and incubated an additional 1.5 h at 60°C. Filters are blotted dry and exposed for autoradiography. If necessary, filters are washed for a third time at 65-68°C and reexposed to film. Other conditions of low stringency which may be used are well known in the art (e. g. , as employed for cross-species hybridizations).

Any technique for mutagenesis known in the art can be used to modify individual nucleotides in a DNA sequence, for purpose of making amino acid substitution (s) in the expressed peptide sequence, or for creating/deleting restriction sites to facilitate further manipulations. Such techniques include but are not limited to, chemical mutagenesis, in vitro site-directed mutagenesis (Hutchinson et al., 1978, J. Biol. Chem. 253: 6551), oligonucleotide-directed mutagenesis (Smith, 1985, Ann. Rev. Genet. 19: 423-463; Hill et al., 1987, Methods Enzymol. 155: 558-568), PCR-based overlap extension (Ho et al., 1989, Gene 77: 51-59), PCR-based megaprimer mutagenesis (Sarkar et al., 1990, Biotechniques 8 : 404-407), etc. Modifications can be confirmed by double stranded dideoxynucleotide DNA sequencing.

In certain embodiments, a nucleic acid encoding a secretory form of a non- secreted HSP is used to practice the methods of the present invention. Such a nucleic acid can be constructed by deleting the coding sequence for the ER retention signal, KDEL.

Optionally, the KDEL coding sequence is replaced with a molecular tag to facilitate the recognition and purification of the HSP, such as the Fc portion of murine IgGl. In another embodiment, a molecular tag can be added to naturally secreted HSPs or oe2M. U. S.

Application Serial No. 09/253, 439 demonstrates that deletion of the ER retention signal of gp96 resulted in the secretion of gp96-Ig peptide-complexes from transfected tumor cells, and the fusion of the KDEL-deleted gp96 with murine IgGl facilitated its detection by

Expression constructs containing cloned HSP or o2M coding sequences can be introduced into the mammalian host cell by a variety of techniques known in the art, including but not limited to calcium phosphate mediated transfection (Wigler et al., 1977, Cell 11: 223-232), liposome-mediated transfection (Schaefer-Ridder et al., 1982, Science 215: 166-168), electroporation (Wolff et al., 1987, Proc. Natl. Acad. Sci. 84: 3344), and microinjection (Cappechi, 1980, Cell 22: 479-488).

Any of the cloning and expression vectors described herein may be synthesized and assembled from known DNA sequences by techniques well known in the art. The regulatory regions and enhancer elements can be of a variety of origins, both natural and synthetic. Some vectors and host cells may be obtained commercially. Non- limiting examples of useful vectors are described in Appendix 5 of Current Protocols in Molecular Biology, 1988, ed. Ausubel et al., Greene Publish. Assoc. & Wiley Interscience, which is incorporated herein by reference ; and the catalogs of commercial suppliers such as Clontech Laboratories, Stratagene Inc. , and Invitrogen, Inc.

Alternatively, number of viral-based expression systems may also be utilized with mammalian cells for recombinant expression of HSPs or a2M. Vectors using DNA virus backbones have been derived from simian virus 40 (SV40) (Hamer et al., 1979, Cell 17: 725), adenovirus (Van Doren et al., 1984, Mol. Cell Biol. 4: 1653), adeno-associated virus (McLaughlin et al., 1988, J. Virol. 62: 1963), and bovine papillomas virus (Zinn et al., 1982, Proc. Natl. Acad. Sci. 79: 4897). In cases where an adenovirus is used as an expression vector, the donor DNA sequence may be ligated to an adenovirus transcription/translation control region, e. g., the late promoter and tripartite leader sequence.

This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e. g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing heterologous products in infected hosts (see, e. g., Logan and Shenk, 1984, Proc. Natl. Acad. Sci. U. S. A.

81 : 3655-3659).

Bovine papillomavirus (BPV) can infect many higher vertebrates, including man, and its DNA replicates as an episome. A number of shuttle vectors have been developed for recombinant gene expression which exist as stable, multicopy (20-300 copies/cell) extrachromosomal elements in mammalian cells. Typically, these vectors contain a segment of BPV DNA (the entire genome or a 69% transforming fragment), a

promoter with a broad host range, a polyadenylation signal, splice signals, a selectable marker, and"poisonless"plasmid sequences that allow the vector to be propagated in E. coli. Following construction and amplification in bacteria, the expression gene construct is transfected into cultured mammalian cells, for example, by the techniques of calcium phosphate coprecipitation or electroporation. For those host cells that do not manifest a transformed phenotype, selection of transformants is achieved by use of a dominant selectable marker, such as histidinol and G418 resistance. For example, BPV vectors such as pBCMGSNeo and pBCMGHis may be used to express HSPs or o2M (Karasuyama et al., Eur. J. Immunol. 18 : 97-104; Ohe et al., Human Gene Therapy 6: 325-33) which may then be transfected into a diverse range of cell types for HSP or o2M expression.

Alternatively, the vaccinia 7. 5K promoter may be used (see, e. g. , Mackett et al., 1982, Proc. Natl. Acad. Sci. U. S. A. 79: 7415-7419; Mackett et al., 1984, J. Virol. 49: 857-864; Panicali et al., 1982, Proc. Natl. Acad. Sci. U. S. A. 79: 4927-4931) In cases where a human host cell is used, vectors based on the Epstein-Barr virus (EBV) origin (OriP) and EBV nuclear antigen 1 (EBNA-1; a trans-acting replication factor) may be used. Such vectors can be used with a broad range of human host cells, e. g., EBO-pCD (Spickofsky et al., 1990, DNA Prot. Eng. Tech. 2: 14-18), pDR2 and DR2 (available from Clontech Laboratories).

Recombinant HSP or o2M expression can also be achieved by a retrovirus- based expression system. In contrast to transfection, retroviruses can efficiently infect and transfer genes to a wide range of cell types including, for example, primary hematopoietic cells. In retroviruses such as Moloney murine leukemia virus, most of the viral gene sequences can be removed and replaced with an HSP or ou2M coding sequence, while the missing viral functions can be supplied in trans. The host range for infection by a retroviral vector can also be manipulated by the choice of envelope used for vector packaging.

For example, a retroviral vector can comprise a 5'long terminal repeat (LTR), a 3'LTR, a packaging signal, a bacterial origin of replication, and a selectable marker. The ND-associated antigenic peptide DNA is inserted into a position between the 5'LTR and 3'LTR, such that transcription from the 5'LTR promoter transcribes the cloned DNA. The 5'LTR comprises a promoter, including but not limited to an LTR promoter, an R region, a U5 region and a primer binding site, in that order. Nucleotide sequences of these LTR elements are well known in the art. A heterologous promoter as

well as multiple drug selection markers may also be included in the expression vector to facilitate selection of infected cells (see McLauchlin et al., 1990, Prog. Nucleic Acid Res. and Molec. Biol. 38: 91-135; Morgenstern et al., 1990, Nucleic Acid Res. 18 : 3587-3596; Choulika et al., 1996, J. Virol 70: 1792-1798; Boesen et al., 1994, Biotherapy 6 : 291-302; Salmons and Gunzberg, 1993, Human Gene Therapy 4 : 129-141 ; and Grossman and Wilson, 1993, Curr. Opin. in Genetics and Devel. 3: 110-114).

The recombinant cells may be cultured under standard conditions of temperature, incubation time, optical density, and media composition. Alternatively, cells may be cultured under conditions emulating the nutritional and physiological requirements of a cell in which the HSP is endogenously expressed. Modified culture conditions and media may be used to enhance production of HSP-peptide complexes. For example, recombinant cells may be grown under conditions that promote inducible HSP expression.

Alpha-2-macroglobulin and HSP polypeptides of the invention may be expressed as fusion proteins to facilitate recovery and purification from the cells in which they are expressed. For example, an HSP or o2M polypeptide may contain a signal sequence leader peptide to direct its translocation across the ER membrane for secretion into culture medium. Further, an HSP or o2M polypeptide may contain an affinity label, such as a affinity label, fused to any portion of the HSP or c2M polypeptide not involved in binding antigenic peptide, such as for example, the carboxyl terminal. The affinity label can be used to facilitate purification of the protein, by binding to an affinity partner molecule.

Various methods for production of such fusion proteins are well known in the art. The manipulations which result in their production can occur at the gene or protein level, preferably at the gene level. For example, the cloned coding region of an HSP or o2M polypeptide may be modified by any of numerous recombinant DNA methods known <BR> <BR> in the art (Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d ed. , Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Ausubel et al., in Chapter 8 of Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, New York). It will be apparent from the following discussion that substitutions, deletions, insertions, or any combination thereof are introduced or combined to arrive at a final nucleotide sequence encoding an HSP or o2M polypeptide.

In various embodiments, fusion proteins comprising the HSP or a2M polypeptide may be made using recombinant DNA techniques. For example, a recombinant gene encoding an HSP or a2M polypeptide may be constructed by introducing an HSP or o2M gene fragment in the proper reading frame into a vector containing the sequence of an affinity label, such that the HSP or a2M polypeptide is expressed as a peptide-tagged fusion protein. Affinity labels, which may be recognized by specific binding partners, may be used for affinity purification of the HSP or o2M polypeptide.

In a preferred embodiment, the affinity label is fused at its amino terminal to the carboxyl terminal of HSP or ou2M. The precise site at which the fusion is made in the carboxyl terminal is not critical. The optimal site can be determined by routine experimentation.

A variety of affinity labels known in the art may be used, such as, but not limited to, the immunoglobulin constant regions, polyhistidine sequence (Petty, 1996, Metal-chelate affinity chromatography, in Current Protocols in Molecular Biology, Vol. 2, Ed. Ausubel et al., Greene Publish. Assoc. & Wiley Interscience), glutathione S-transferase (GST; Smith, 1993, Methods Mol. Cell Bio. 4: 220-229), the E. coli maltose binding protein (Guan et al., 1987, Gene 67: 21-30), and various cellulose binding domains (U. S. Patent Nos. 5,496, 934; 5,202, 247; 5,137, 819; Tomme et al., 1994, Protein Eng. 7: 117-123), etc.

Other affinity labels may impart fluorescent properties to an HSP or o2M polypeptide, e. g., portions of green fluorescent protein and the like. Other possible affinity labels are short amino acid sequences to which monoclonal antibodies are available, such as but not limited to the following well known examples, the FLAG epitope, the myc epitope at amino acids 408-439, the influenza virus hemagglutinin (HA) epitope. Other affinity labels are recognized by specific binding partners and thus facilitate isolation by affinity binding to the binding partner which can be immobilized onto a solid support. Some affinity labels may afford the HSP or o2M polypeptide novel structural properties, such as the ability to form multimers. Dimerization of an HSP or o2M polypeptide with a bound peptide may increase avidity of interaction between the HSP or o2M polypeptide and its partner in the course of antigen presentation. These affinity labels are usually derived from proteins that normally exist as homopolymers. Affinity labels such as the extracellular domains of CD8 (Shiue et al., 1988, J. Exp. Med. 168: 1993-2005), or CD28 (Lee et al., 1990, J. Immunol.

145: 344-352), or portions of the immunoglobulin molecule containing sites for interchain

disulfide bonds, could lead to the formation of multimers. As will be appreciated by those skilled in the art, many methods can be used to obtain the coding region of the above- mentioned affinity labels, including but not limited to, DNA cloning, DNA amplification, and synthetic methods. Some of the affinity labels and reagents for their detection and isolation are available commercially.

A preferred affinity label is a non-variable portion of the immunoglobulin molecule. Typically, such portions comprise at least a functionally operative CH2 and CH3 domain of the constant region of an immunoglobulin heavy chain. Fusions are also made using the carboxyl terminus of the Fc portion of a constant domain, or a region immediately amino-terminal to the CH1 of the heavy or light chain. Suitable immunoglobulin-based affinity label may be obtained from IgG-1,-2,-3, or-4 subtypes, IgA, IgE, IgD, or IgM, but preferably IgGl. Preferably, a human immunoglobulin is used when the HSP or o2M polypeptide is intended for in vivo use for humans. Many DNA encoding immunoglobulin light or heavy chain constant regions is known or readily available from cDNA libraries.

See, for example, Adams et al., Biochemistry, 1980,19 : 2711-2719; Gough et al., 1980, <BR> <BR> Biochemistry, 19: 2702-2710; Dolby et al., 1980, Proc. Natl. Acad. Sci. U. S. A. , 77: 6027-<BR> 6031 ; Rice et al., 1982, Proc. Natl. Acad. Sci. U. S. A. , 79: 7862-7865; Falkner et al., 1982, Nature, 298: 286-288; and Morrison et al., 1984, Ann. Rev. Immunol, 2: 239-256. Because many immunological reagents and labeling systems are available for the detection of immunoglobulins, the HSP or o2M polypeptide-Ig fusion protein can readily be detected and quantified by a variety of immunological techniques known in the art, such as the use of enzyme-linked immunosorbent assay (ELISA), immunoprecipitation, fluorescence activated cell sorting (FACS), etc. Similarly, if the affinity label is an epitope with readily available antibodies, such reagents can be used with the techniques mentioned above to detect, quantitate, and isolate the HSP or o2M polypeptide containing the affinity label. In many instances, there is no need to develop specific antibodies to the HSP or o2M polypeptide.

A particularly preferred embodiment is a fusion of an HSP or oe2M polypeptide to the hinge, the CH2 and CH3 domains of human immunoglobulin G-1 (IgG- 1; see Bowen et al., 1996, J. Immunol. 156: 442-49). This hinge region contains three cysteine residues which are normally involved in disulfide bonding with other cystines in the Ig molecule. Since none of the cysteines are required for the peptide to function as a

tag, one or more of these cysteine residues may optionally be substituted by another amino acid residue, such as for example, serine.

Various leader sequences known in the art can be used for the efficient secretion of HSP or o2M polypeptide from bacterial and mammalian cells (von Heijne, 1985, J. Mol. Biol. 184: 99-105). Leader peptides are selected based on the intended host cell, and may include bacterial, yeast, viral, animal, and mammalian sequences. For example, the herpes virus glycoprotein D leader peptide is suitable for use in a variety of mammalian cells. A preferred leader peptide for use in mammalian cells can be obtained from the V-J2-C region of the mouse immunoglobulin kappa chain (Bernard et al., 1981, Proc. Natl. Acad. Sci. 78 : 5812-5816). Preferred leader sequences for targeting HSP or o2M polypeptide expression in bacterial cells include, but are not limited to, the leader sequences of the E. coli proteins OmpA (Hobom et al., 1995, Dev. Biol. Stand. 84: 255-262), Pho A (Oka et al., 1985, Proc. Natl. Acad. Sci 82: 7212-16), OmpT (Johnson et al., 1996, Protein Expression 7: 104-113), LamB and OmpF (Hoffinan & Wright, 1985, Proc. Natl. Acad. Sci.

USA 82 : 5107-5111), ß-lactamase (Kadonaga et al., 1984, J. Biol. Chem. 259: 2149-54), enterotoxins (Morioka-Fujimoto et al., 1991, J. Biol. Chem. 266: 1728-32), and the Staphylococcus aureus protein A (Abrahmsen et al., 1986, Nucleic Acids Res. 14: 7487- 7500), and the B. subtilis endoglucanase (Lo et al., Appl. Environ. Microbiol. 54: 2287- 2292), as well as artificial and synthetic signal sequences (Maclntyre et al., 1990, Mol. Gen.

Genet. 221: 466-74; Kaiser et al., 1987, Science, 235: 312-317).

DNA sequences encoding a desired affinity label or leader peptide, which may be readily obtained from libraries, produced synthetically, or may be available from commercial suppliers, are suitable for the practice of this invention. Such methods are well known in the art.

5.3 COMPLEXING PEPTIDE FRAGMENTS TO HSP AND o2M Described herein are methods for complexing in vitro the HSP or a2M with a population of peptides which have been generated by digestion of a protein preparation of antigenic cells. The complexing reaction can result in the formation of a covalent bond between a HSP and a peptide, or a a2M and a peptide. The complexing reaction can also result in the formation of a non-covalent association between a HSP and a peptide, or a c2M and a peptide.

In a method which produces non-covalent HSP-antigenic molecule complexes and o2M-antigenic molecule complexes, a complex is prepared according to the method described by Blachere et al., 1997 J. Exp. Med. 186 (8): 1315-22, which incorporated by reference herein in its entirety. Blachere teaches in vitro complexing of hsps to antigenic molecule. The protocol described in Blachere can be modified such that the hsp component is substituted by o2M. Binder et al. (2001, J. Immunol. 166: 4968-72) demonstrates that the Blachere method yields complexes of o2M bound to antigenic molecules.

Prior to complexing, the HSPs can be pretreated with ATP or low pH to remove any peptides that may be non-covalently associated with the HSP of interest. When the ATP procedure is used, excess ATP is removed from the preparation by the addition of apyranase as described by Levy, et al., 1991, Cell 67 : 265-274. When the low pH procedure is used, the buffer is readjusted to neutral pH by the addition of pH modifying reagents. A preferred, exemplary protocol for the complexing of a population of peptides (average length between 7 to 20 amino acids) to an HSP or o2M in vitro is discussed below The population of peptides (Ittg) and the pretreated HSP (9/. Lg) are admixed to give an approximately 5 peptides: 1 stress protein molar ratio. Then, the mixture is incubated for 15 minutes to 3 hours at 4° to 45°C in a suitable binding buffer such as one containing 20mM sodium phosphate, pH 7.2, 350mM NaCI, 3mM MgCl2 and ImM phenyl methyl sulfonyl fluoride (PMSF). The preparations are centrifuged through a Centricon 10 assembly (Millipore) to remove any unbound peptide. The non-covalent association of the peptides with the stress proteins can be assayed by High Performance Liquid Chromatography (HPLC) or Mass Spectrometry (MS).

In an alternative embodiment of the invention, preferred for producing non- covalent complexes of HSP70 to peptide fragments: 5-10 micrograms of purified HSP70 is incubated with equimolar quantities of peptide fragments in 20mM sodium phosphate buffer pH 7.5, 0. 5M NaCI, 3mM MgCl2 and 1mM ADP in a volume of 100 microliter at 37°C for 1 hr. This incubation mixture is centrifuged one or more times if necessary, through a Centricon 10 assembly (Millipore) to remove any unbound peptide.

In an alternative embodiment of the invention, preferred for producing complexes of gp96 or HSP90 to peptide fragments, 5-10 micrograms of purified gp96 or HSP90 is incubated with equimolar or excess quantities of the peptide fragments in a

suitable buffer such as one containing 20mM sodium phosphate buffer pH 7.5, 0. 5M NaCl, 3nM MgC12 at 60-65°C for 5-20 min. This incubation mixture is allowed to cool to room temperature and centrifuged one or more times if necessary, through a Centricon 10 assembly (Millipore) to remove any unbound peptide.

Following complexing, an immunogenic HSP-peptide complex or oe2M- peptide complex can optionally be assayed using for example the mixed lymphocyte target cell assay (MLTC) described below. Once HSP-peptide complexes have been isolated and diluted, they can be optionally characterized further in animal models using the preferred administration protocols and excipients discussed below.

As an alternative to making non-covalent complexes of HSPs and peptides, a population of peptides can be covalently attached to HSPs. Covalently linked complexes are the complexes of choice when a B cell response is desired.

In one embodiment, HSPs are covalently coupled to peptide fragments by chemical crosslinking. Chemical crosslinking methods are well known in the art. For example, in a preferred embodiment, glutaraldehyde crosslinking may be used.

Glutaradehyde crosslinking has been used for formation of covalent complexes of peptides and HSPs (see Barrios et al., 1992, Eur. J. Immunol. 22: 1365-1372). Preferably, 1-2 mg of HSP-peptide complex is crosslinked in the presence of 0.002% glutaraldehyde for 2 hours. Glutaraldehyde is removed by dialysis against phosphate buffered saline (PBS) overnight (Lussow et al., 1991, Eur. J. Immunol. 21: 2297-2302). Alternatively, a HSP and a population of peptides can be crosslinked by ultraviolet (UV) crosslinking under conditions known in the art.

In another embodiment of the invention, a population of peptides can be complexed to o2M by incubating the peptide fragments with c2M at a 50: 1 molar ratio and incubated at 50° C for 10 minutes followed by a 30 minute incubation at 25°C. Free (uncomplexed) peptides are then removed by size exclusion filters. Protein-peptide complexes are preferably measured by a scintillation counter to make sure that on a per molar basis, each protein is observed to bind equivalent amounts of peptide (approximately 0.1% of the starting amount of the peptide). For details, see Binder, 2001, J. Immunol.

166 (8): 4968-72, which is incorporated herein by reference in its entirety.

Alternatively, a population of antigenic peptides can be complexed to of 2M covalently by methods as described in PCT publications WO 94/14976 and WO 99/50303

for complexing a peptide to o2M, which are incorporated herein by reference in their entirety. Covalent linking of a population of antigenic peptides to o2M can be performed using a bifunctional crosslinking agent. Such crosslinking agents and methods of their use are also well known in the art.

5.4 VACCINES THAT CAN BE USED The vaccines that can be used with the HSP or o2M preparations of the invention include but are not limited to live vaccines, inactivated vaccines, attenuated vaccines, subunit vaccines, and nucleic acid-based vaccines. Subunit vaccines may be multivalent or univalent, and may, for example, contain purified pathogen antigens, such as isolated viral coat proteins, and bacterial cell wall molecules, etc. Multivalent vaccines are made from recombinant viruses that direct the expression of more than one antigen. Until recently, vaccines are typically used for prophylaxis against infectious diseases. However, vaccines based on tumor antigens, e. g. , containing tumor specific or tumor-associated antigens, have been developed for the treatment or prevention of various types of cancers.

Non-limiting examples of tumor antigens that can be used in a vaccine composition may include KS 1/4 pan-carcinoma antigen (Perez and Walker, 1990, J. Immunol. 142: 3662- 3667; Bumal, 1988, Hybridoma 7 (4): 407-415); ovarian carcinoma antigen (CA125) (Yu, et al. , 1991, Cancer Res. 51 (2): 468-475) ; prostatic acid phosphate (Tailer, et al. , 1990, Nucl.

Acids Res. 18 (16): 4928); prostate specific antigen (Henttu and Vihko, 1989, Biochem.

Biophys. Res. Comm. 160 (2): 903-910; Israeli, et al. , 1993, Cancer Res. 53: 227-230);<BR> melanoma-associated antigen p97 (Estin, et al. , 1989, J. Natl. Cancer Inst. 81 (6): 445-446);<BR> melanoma antigen gp75 (Vijayasardahl, et al. , 1990, J. Exp. Med. 171 (4): 1375-1380); high<BR> molecular weight melanoma antigen (Natali, et al. , 1987, Cancer 59: 55-63), the MAGE<BR> family of antigens (Hu et al. , 1996, Cancer Res. 56: 2479-2483; Marchand et al. , 1995, Int.

J. Cancer 63: 883-885) and prostate specific membrane antigen. The HSP and o2M preparations of the invention can also be used with such cancer vaccines. The cancer vaccines that can be used with the methods of invention are reviewed in various publications, e. g. , Pardoll, 2000, Clin. Immunol. 95 (1 Pt 2): S44-62 and Stevenson, 1999, Ann Oncol. 10: 1413-8 the contents of which are incorporated herein by reference in their entireties.

Many methods may be used to introduce the vaccine; these include but are not limited to oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous,

intranasal routes, and via scarification (scratching through the top layers of skin, e. g., using a bifurcated needle).

The patient to which the vaccine is administered is preferably a mammal, most preferably a human, but can also be a non-human animal including but not limited to primates, cows, horses, sheep, pigs, fowl (e. g., chickens), goats, cats, dogs, hamsters, mice and rats.

Examples of vaccine compositions that can be used with the Hsp and o2M preparations of the invention include but are not limited to bacillus Calmette-Guerin vaccine, brucella strain 19 vaccine, cholera vaccine, diphtheria-tetanus toxoids-petussis vaccines, foot-and-mouth-disease vaccine, Haffkine's vaccines, various hepatitis virus vaccines, human diploid cell rabies virus, poliovirus vaccine, influenza virus vaccine, measles vaccine, measles-mumps-rubella vaccine, plague vaccine, pneumococcal vaccine, rickettsia vaccine, Sabin vaccine, Semple vaccine, smallpox vaccine, staphylococcus vaccine, typhoid vaccine, typhus vaccine, whooping cough vaccine, and yellow fever vaccine. The vaccines that can be used with the methods of invention are reviewed in various publications, e. g., The Jordan Report 2000, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, United States, the content of which is incorporated herein by reference in its entirety.

The vaccine composition may comprise adjuvants, or may be administered together with one or more adjuvants. Adjuvants that can be used include but are not limited to mineral salt adjuvants or mineral salt gel adjuvants, particulate adjuvants, microparticulate adjuvants, mucosal adjuvants, and immunostimulatory adjuvants.

Examples of adjuvants include, but are not limited to, aluminum hydroxide, aluminum phosphate gel, Freund's Complete Adjuvant, Freund's Incomplete Adjuvant, squalene or squalane oil-in-water adjuvant formulations, biodegradable and biocompatible polyesters, polymerized liposomes, triterpenoid glycosides or saponins (e. g., QuilA and QS-21, also sold under the trademark STIMULON, ISCOPREP), N-acetyl-muramyl-L-threonyl-D- isoglutamine (Threonyl-MDP, sold under the trademark TERMURTIDE), LPS, monophosphoryl Lipid A (3D-MLAsold under the trademark MPL).

5.5 KITS, DOSAGE REGIMENS, ADMINISTRATION AND FORMULATIONS

Kits are also provided for carrying out the vaccination methods of the present invention. In a specific embodiment, a kit comprises a first container containing a heat shock protein preparation or an o2M preparation in an amount effective to increase an immune response elicited by a vaccine composition against a component of the vaccine composition against which an immune response is desired; and a second container containing the vaccine composition in an amount that, when administered before, concurrently with, or after the administration of the heat shock protein preparation or the o2M preparation in the first container, is effective to induce an immune response against the component. In an alternate embodiment, the kit comprises a container containing both the HSP preparation and the vaccine composition. In another alternate embodiment, the kit comprises a container containing both the o2M preparation and the vaccine composition. In a specific embodiment, the kit comprises a first and second container, the vaccine composition does not comprise an HSP or an a2M. In another specific embodiment, when the kit comprises a container containing both the HSP/o2M, and the vaccine composition does not comprise an HSP or an o2M.

Kits of the invention are provided that comprise in a container a vaccine composition in an amount effective to treat or prevent a disease or disorder; and in another container either a heat shock protein preparation or an a2M preparation in an amount effective to increase or boost an immune response elicited by the vaccine. In an embodiment, the amount of vaccine composition present in the container is insufficient for inducing an immune response in a subject if administered independent of the heat shock protein preparation or of the o2M preparation in the other container. The kit may optionally be accompanied by instructions.

The dosage of HSP preparation or o2M preparation to be administered depends to a large extent on the condition and size of the subject being treated as well as the amount of vaccine composition administered, the frequency of treatment and the route of administration. Regimens for continuing therapy, including site, dose and frequency may be guided by the initial response and clinical judgment.

Depending on the route of administration and the type of HSPs in the HSP preparation, the amount of HSP in the HSP preparation can range, for example, from 0.1 to 1000 zig per administration. The preferred amounts of gp96 or hsp70 are in the range of 10 to 600 it per administration and 0.1 to 10 ju. g if the HSP preparation is administered

intradermally. For hsp 90, the preferred amounts are about 50 to 1000, ug per administration, and about 5 to 50, Ag for intradermal administration. The amount of a2M administered can range from 2 to 1000 ug, preferably 20 to 500, ug, most preferably about 25 to 250/g, given once weekly for about 4-6 weeks, intradermally with the site of administration varied sequentially.

In one preferred embodiment, the HSP preparation or the c2M preparation is administered concurrently with the administration of a vaccine. Concurrent administration of an HSP preparation or a2M preparation and a vaccine means that the HSP or o2M preparation is given at reasonably the same time as the vaccine. This method provides that the two administrations are performed within a time frame of less than one minute to about five minutes, or up to about sixty minutes from each other, for example, at the same doctor's visit.

Because of the administration of the HSP preparation or the a2M preparation, lesser amount of vaccine is required to elicit an immune respone in a subject.

In specific embodiments, a reduction of about 10%, 20%, 30%, 40% and 50% of the amount of vaccine composition can be achieved. Even sub-immunogenic amounts of the vaccine composition can be used provided that an appropriate amount of the HSP preparation or o2M preparation is used in conjunction. The amount of vaccine composition to be used with a HSP preparation or o2M preparation, including amounts in the sub- immunogenic range, can be determined by dose-response experiments conducted in animal models by methods well known in the art.

Solubility and the site of the vaccination are factors which should be considered when choosing the route of administration of the HSP or o2M preparation of the invention. The mode of administration can be varied, including, but not limited to, e. g., subcutaneously, intravenously, intraperitoneally, intramuscularly, intradermally or mucosally. Mucosal routes can further take the form of oral, rectal and nasal administration. With the above factors taken into account, it is preferable to administer the HSP or the o2M to a site that is the same or proximal to the site of vaccination.

In an embodiment of the invention, HSPs or o2M may be administered using any desired route of administration. Advantages of intradermal administration include use of lower doses and rapid absorption, respectively. Advantages of subcutaneous or intramuscular administration include suitability for some insoluble suspensions and oily

suspensions, respectively. Mucosal routes of administration include, but are not limited to, oral, rectal and nasal administration. Preparations for mucosal administrations are suitable in various formulations as described below.

In a preferred embodiment, the invention provides for a method of introducing an HSP preparation including, but not limited to, hsp70, hsp90 and gp96 alone or in combination with each other into a subject concurrently with the administration of a vaccine at the same site or at a site in close proximity. In another preferred embodiment, the invention provides for a method of introducing an o2M preparation concurrently with the administration of a vaccine at the same site or at a site in close proximity. Preferably the HSP preparation or the o2M preparation are not administered with the vaccine composition in admixture.

If the HSP or o2M preparation is water-soluble, then it may be formulated in an appropriate buffer, for example, phosphate buffered saline or other physiologically compatible solutions, preferably sterile. Alternatively, if the resulting complex has poor solubility in aqueous solvents, then it may be formulated with a non-ionic surfactant such as Tween, or polyethylene glycol. Thus, the compounds and their physiologically acceptable solvates may be formulated for administration by inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral, or rectal administration or, in the case of tumors, directly injected into a solid tumor.

For oral administration, the pharmaceutical preparation may be in liquid form, for example, solutions, syrups or suspensions, or may be presented as a drug product for reconstitution with water or other suitable vehicle before use. Such a liquid preparation may be prepared by conventional means with pharmaceutically acceptable additives such as <BR> <BR> suspending agents (e. g. , sorbitol syrup, cellulose derivatives or hydrogenated edible fats);<BR> emulsifying agents (e. g. , lecithin or acacia); non-aqueous vehicles (e. g. , almond oil, oily<BR> esters, or fractionated vegetable oils); and preservatives (e. g. , methyl or propyl-p- hydroxybenzoates or sorbic acid). The pharmaceutical preparation may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically <BR> <BR> acceptable excipients such as binding agents (e. g. , pregelatinized maize starch, polyvinyl<BR> pyrrolidone or hydroxypropyl methylcellulose); fillers (e. g. , lactose, microcrystalline<BR> cellulose or calcium hydrogen phosphate); lubricants (e. g. , magnesium stearate, talc or

silica); disintegrants (e. g. , potato starch or sodium starch glycolate); or wetting agents (e. g., sodium lauryl sulphate). The tablets may be coated by methods well-known in the art.

The HSP preparation for oral administration may be suitably formulated to give controlled release of the active compound.

For buccal administration, the preparation may take the form of tablets or lozenges formulated in conventional manner.

The preparation may be formulated for parenteral administration by <BR> <BR> injection, e. g. , by bolus injection or continuous infusion. Formulations for injection may be<BR> presented in unit dosage form, e. g. , in ampoules or in multi-dose containers, with an added preservative. The preparation may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e. g. , sterile pyrogen-free water, before use.

The preparation may also be formulated in a rectal preparation such as a suppository or retention enema, e. g. , containing conventional suppository bases such as cocoa butter or other glycerides.

In addition to the formulations described previously, the preparation may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the preparation may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophilic drugs.

For administration by inhalation, the preparation for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e. g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, <BR> <BR> e. g. , gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.

The preparation may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the HSP preparation or the a2M preparation. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.

5.6 ACTIVATION AND ADMINISTRATION OF ANTIGEN-PRESENTING CELLS APC can be obtained, maintained and/or expanded by any of various methods known in the art. In one embodiment, the antigen-presenting cells, including but not limited to macrophages, dendritic cells and B-cells, can be obtained by production in vitro from stem and progenitor cells from human peripheral blood or bone marrow as described by Inaba, K., et al. , 1992, J. Exp. Med. 176: 1693-1702. In another embodiment, human macrophages are used, obtained from human blood cells. By way of example but not limitation, macrophages can be obtained as follows: Mononuclear cells are isolated from peripheral blood of a patient (preferably the patient to be treated), by Ficoll-Hypaque gradient centrifugation.

Tissue culture dishes are pre-coated with the patient's own serum or with other AB+ human serum and incubated at 37°C for 1 hr. Non-adherent cells are removed by pipetting. To the adherent cells left in the dish, is added cold (4°C) 1mM EDTA in phosphate-buffered saline and the dishes are left at room temperature for 15 minutes. The cells are harvested, washed with RPMI buffer and suspended in RPMI buffer. Increased numbers of macrophages can be obtained by incubating at 37°C with macrophage-colony stimulating factor (M-CSF). In a preferred embodiment, increased numbers of dendritic cells can be obtained by incubating with granulocyte-macrophage-colony stimulating factor (GM-CSF) as described in detail by Inaba, K. , et al. , 1992, J. Exp. Med. 176: 1693-1702.

5.6. 1 Activation of Antigen Presenting Cells With HSP or o2M Preparations APC can be activated with an HSP or o2M preparation of the invention by incubating the cells in vitro with the complexes. Preferably, the APC are activated with a HSP preparation or a2M preparation by incubating in vitro with the hsp-complex or o2M- complex at 37°C for 15 minutes to 24 hours. By way of example but not limitation, 4x107 macrophages can be incubated with 10 microgram gp96 per ml or 100 microgram hsp90 per

ml at 37°C for 15 minutes to 24 hours in 1 ml plain RPMI medium. The cells are washed three times and resuspended in a physiological medium preferably sterile, at a convenient concentration (e. g., 1XI07/Ml) for infusion in a patient. Preferably, the patient into which the sensitized APCs are infused is the patient from which the APC were originally isolated (autologous embodiment).

5.6. 2 Reinfusion of Activated APC The activated macrophages and other APC can be reinfused into the subject by conventional clinical procedures, such as but not limited to intravenous, subcutaneous, intradermal, and intraperitoneal administration. These activated cells are reinfused, preferentially by systemic administration into the autologous patient. Subjects generally receive from about 106 to about 1012 sensitized macrophages, depending on the condition of the subject.

5.7 DETERMINATION OF IMMUNOGENICITY OF VACCINES AFTER HSP TREATMENT In an optional procedure, the production of or increase in immunogenicity of a vaccine that is used with the HSP or a2M preparation of the invention can be assessed using various methods well known in the art.

In one method, the immunogenicity of the vaccine and HSP preparation or o2M preparation is determined by measuring antibodies produced in response, by an antibody assay, such as an enzyme-linked immunosorbent assay (ELISA) assay. Methods for such assays are well known in the art (see, e. g., Section 2.1 of Current Protocols in Immunology, Coligan et aL (eds. ), John Wiley and Sons, Inc. 1997). For example, microtitre plates (96-well Immune Plate II, Nunc) are coated with 50 jul/well of a 0.75 yg/ml extract or lysate of a cancer cell or infected cell in PBS at 4°C for 16 hours and at 20°C for 1 hour. The wells are emptied and blocked with 200 ul PBS-T-BSA (PBS containing 0.05% (v/v) TWEEN 20 and 1% (w/v) bovine serum albumin) per well at 20°C for 1 hour, then washed 3 times with PBS-T. Fifty ul/well of plasma or CSF from a vaccinated animal (such as a model mouse or a human patient with or without administration of a HSP preparation) is applied at 20°C for 1 hour, and the plates are washed 3 times with PBS-T. The antigen antibody activity is then measured colorimetrically after incubating at 20°C for 1 hour with 50/well of sheep anti-mouse or anti-human immunoglobulin, as appropriate, conjugated with horseradish peroxidase

diluted 1: 1,500 in PBS-T-BSA and (after 3 further PBS-T washes as above) with 50 fil of an <BR> <BR> o-phenylene diamine (OPD) -H202 substrate solution. The reaction is stopped with 150, ul of 2M H2S04 after 5 minutes and absorbance is determined in a photometer at 492 nm (ref.

620 nm), using standard techniques.

In another method, the"tetramer staining"assay (Altman et al. , 1996, Science 274: 94-96) may be used to identify antigen-specific T-cells. For example, in one embodiment, an MHC molecule containing a specific peptide antigen, such as a tumor- specific antigen, is multimerized to make soluble peptide tetramers and labeled, for example, by complexing to streptavidin. The MHC-peptide antigen complex is then mixed with a population of T cells obtained from a patient treated with a vaccine and the HSP preparation. Biotin is then used to stain T cells which express the tumor-specific antigen of interest.

Furthermore, using the mixed lymphocyte target culture assay, the cytotoxicity of T cells can be tested in a 4 hour 5'Cr-release assay (see Palladino et al., 1987, Cancer Res. 47: 5074-5079). In this assay, the mixed lymphocyte culture is added to a target cell suspension to give different effector: target (E: T) ratios (usually 1: 1 to 40: 1). The target cells are pre-labeled by incubating 1x106 target cells in culture medium containing 500 yCi of slCr per ml for one hour at 37°C. The cells are washed three times following labeling. Each assay point (E: T ratio) is performed in triplicate and the appropriate controls incorporated to measure spontaneous Cr release (no lymphocytes added to assay) and 100% release (cells lysed with detergent). After incubating the cell mixtures for 4 hours, the cells are pelleted by centrifugation at 200g for 5 minutes. The amount of 51 Cr released into the supernatant is measured by a gamma counter. The percent cytotoxicity is measured as cpm in the test sample minus spontaneously released cpm divided by the total detergent released cpm minus spontaneously released cpm. In order to block the MHC class I cascade a concentrated hybridoma supernatant derived from K-44 hybridoma cells (an anti- MHC class I hybridoma) is added to the test samples to a final concentration of 12.5%.

Alternatively, the ELISPOT assay can be used to measure cytokine release in vitro by cytotoxic T cells after stimulation with vaccine and HSP preparation or a2M preparation. Cytokine release is detected by antibodies which are specific for a particular cytokine, such as interleukin-2, tumor necrosis factor a or interferon- (for example, see Scheibenbogen et al., 1997, Int. J. Cancer, 71: 932-936). The assay is carried out in a

microtitre plate which has been pre-coated with an antibody specific for a cytokine of interest which captures the cytokine secreted by T cells. After incubation of T cells for 24- 48 hours in the coated wells, the cytotoxic T cells are removed and replaced with a second labeled antibody that recognizes a different epitope on the cytokine. After extensive washing to remove unbound antibody, an enzyme substrate which produces a colored reaction product is added to the plate. The number of cytokine-producing cells is counted under a microscope. This method has the advantages of short assay time, and sensitivity without the need of a large number of cytotoxic T cells.

5. 8 TREATMENT AND PREVENTION OF INFECTIOUS DISEASES Infectious diseases that can be treated or prevented by use of a vaccine composition in conjunction with the methods of the present invention are caused by infectious agents including, but not limited to, viruses, bacteria, fungi protozoa and parasites. Some of the commonly-used vaccine compositions against infectious diseases are described in Section 5.2. Other examples are described in The Jordan Report 2000, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, United States, the content of which is incorporated herein by reference in its entirety.

Viral diseases that can be treated or prevented by use of a vaccine composition in conjunction with the methods of the present invention include, but are not limited to, those caused by hepatitis A virus, hepatitis B virus, hepatitis C virus, influenza, varicella, adenovirus, herpes simplex I virus, herpes simplex II virus, rinderpest, rhinovirus, echovirus, rotavirus, respiratory syncytial virus, papilloma virus, papova virus, cytomegalovirus, echinovirus, arbovirus, hantavirus, coxsackie virus, mumps virus, measles virus, rubella virus, polio virus, human immunodeficiency virus type I (HIV-I), and human immunodeficiency virus type II (HIV-II).

Bacterial diseases that can be treated or prevented by use of a vaccine composition in conjunction with the methods of the present invention are caused by bacteria including, but not limited to, mycobacteria rickettsia, mycoplasma, neisseria and legionella.

Protozoal diseases that can be treated or prevented by use of a vaccine composition in conjunction with the methods of the present invention are caused by protozoa including, but not limited to, leishmania, kokzidioa, and trypanosoma.

Parasitic diseases that can be treated or prevented by use of a vaccine composition in conjunction with the methods of the present invention are caused by parasites including, but not limited to, chlamydia and rickettsia.

5.9 TREATMENT OF CANCER A number of cancer vaccines for treatment of melanoma, pancreatic carcinoma, breast cancer, prostate cancer are currently in clinical trials. The HSP or o2M preparation can be used in conjunction with such cancer vaccines for the treatment and prevention of the respective types of cancers. Examples of cancer vaccines that can be used with the methods of invention are described in various publications, e. g. , Pardoll, 2000, Clin. Immunol. 95 (1 Pt 2) : S44-62 and Stevenson, 1999, Ann Oncol. 10: 1413-8. Cancers that can also be treated by use of a vaccine composition in conjunction with the methods of the present invention include, but are not limited to the following types of cancer: human sarcomas and carcinomas, e. g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms'tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma; leukemias, e. g., acute lymphocytic leukemia and acute myelocytic leukemia (myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); and polycythemia vera, lymphom (Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, and heavy chain disease.

6. EXAMPLE: HEAT SHOCK PROTEIN ACTIVATES ANTIGEN PRESENTING CELLS

Antigen presenting cells (APCs), such as dendritic cells, B cells, and macrophages, are key components of innate and adaptive immune responses. They are normally quiescent and require activation for their function. The identity of signals which activate APCs is thus a crucial question. Apparently, necrotic but not apoptotic cell death leads to release of HSPs. Applicant's experimentation with CD1 lb+ cells as shown in this section reveals that increased concentration of HSPs in the extracellular milieu where there are APCs induces secretion of cytokines and upregulates surface expression of antigen- presenting and co-stimulatory molecules, including, but not limited to, B7-1, B7-2, and MHC class II. HSPs interact with these APCs through the conserved NFKB pathway.

During antigen presentation, the complementary ligands on the T cells that associate with B7-1, B7-2, and MHC class II are CD28, CD28, and T-cell antigen surface receptors (TCRs) with CD4, respectively (See Banchereau, 1998, Nature 392: 247). Thus, HSPs have been identified to constitute a signal for APC activation.

6.1 MATERIALS AND METHODS HSPs, and Antibodies. Hsp90, hsp70 and gp96 were purified simultaneously from C57BL/6 mouse liver as described (8). Antibodies against CD80 (B7-1), CD86 (B7- 2), CD40, CD1 lb, and MHC II for FACS analysis were purchased from Pharmingen, San Diego, CA.

Assay of LPS content. The LPS content was measured by the LAL assay (LAL Kit QCL-1000, BIOWHITTAKER, Walkersville, Maryland).

Preparation of necrotic and apoptotic cells. Cells were frozen and thawed through four cycles of liquid nitrogen-room temperature treatments, in order to mimic necrosis. Cells were irradiated (7,500 rads) in order to initiate apoptosis.

Generation of bone marrow derived DCs. Femurs and tibia of C57BL/6 mice were removed. The marrow was flushed out from the bones with media and leukocytes obtained were cultured as described (Lutz et al. , 1999, J. Immunol. Methods, 223: 77-92).

Cytokine assay. Cells (5x104) were incubated for 20 hours at 37°C in complete medium with 5% fetal calf serum, or with increasing quantities of HSPs, in 96 well, flat bottom plates. Supernatants were harvested and assayed by ELISA for TNF-ot, IL12, IL-lj8, GM-CSF and Interferon-, (IFN-y). IL-lß, TNF-ot, GM-CSF and IFN-kits

were purchased from Endogen Inc., Woburn, MA, IL12 kit was purchased from R&D Systems, Inc. , Minneapolis, MN.

Preparation of nuclear extracts and Electrophoretic mobility shift assay.

APCs were washed with PBS (LPS-free) and re-suspended in cold lysis buffer [buffer A: lOmM Hepes (pH 7.9), lOmM KC1, 0. 1mM EDTA, O. 1mM EGTA, ImM DTT, 0. 5mM PMSF, l, uM aprotinin, l, uM pepstatin and 14juM leupeptin] with 0. 1% NP40 and incubated on ice for 30 min. Nuclei were pelleted at 14000 rpm for 2 min at 4°C. Supernatant was collected and protein concentration was measured by Bradford assay. The standard DNA- binding reaction was performed using KB DNA probe (5'- AGTTGAGGGGACTTTCCCAGGC-3'), as described by Dignam et al. (1983, Nucleic Acids Res. 11: 1475-89).

6.2 NECROTIC BUT NOT APOPTOTIC CELLS RELEASE HSPs.

Cell death can be achieved in a variety of ways, popularly classified into two : apoptotic and necrotic. The inventors investigated which of these two forms of death can result in release of the major HSPs: hsp70, hsp90, calreticulin (CRT) and gp96. E. G7 cells were subjected to a freeze-thaw procedure as a necrosis-mimetic or were irradiated as a form of apoptosis-mimetic process, as described in Methods. Cells were checked for necrosis visually under the microscope, and for apoptosis, by externalization of phosphatidyl serine (as detected by staining with Annexin V) and degradation of PARP by caspases (Schletter et al. , 1995, Arch. Microbiol. 164: 383-9). The supernatants of the treated cells were collected immediately after treatment or 24 hours after treatment by either method and analyzed by SDS-PAGE and immunoblotting with antibodies to the 4 HSPs. It was observed that necrotic but not apoptotic death led to release of all four HSPs. No Hsps were detected in the supematants of apoptotic cells even 24 hours after death.

6.3 ACTIVATION OF CD1 lb+ CELLS BY HEAT SHOCK PROTEINS Three representative HSPs, hsp90 and gp96 (of the hsp90 family) and hsp70 were tested for their abilities to activate antigen presenting cells. Hsp90 and hsp70 are cytosolic proteins whereas gp96 is localized in the endoplasmic reticulum. Altogether, the three HSPs constitute the most abundant soluble components (>2 % of the total protein) of the mammalian cells. Approximately 30 mg gp96, 200 tug hsp70 and 400 jUg hsp90 can be isolated in purified form from 2-5 x 108 cells. The three HSPs were purified from livers of C57BL/6 mice as described below were shown to be homogenous by SDS-polyacrylamide

gel electrophoresis (Fig. 1A) and were identified by immunoblotting with respective monoclonal antibodies. Peritoneal cells from naive mice or mice previously injected intraperitoneally with pristane were positively selected for CD 1 lb+ cells as described, which were then cultured in vitro with increasing quantities of gp96 for 20 hours at 37°C.

Supernatants were harvested and tested for the presence of IL-lß, TNF-ot, GM-CSF, IL-12 and interferon-y (as a negative control) by ELISA (Fig. 1B). Treatment with anti-CD 1 lb antibody during or after positive selection did not result in activation of cells. Gp96 was found to activate, in a titratable manner, secretion of all the cytokines tested, except interferon-'y. Similar results were obtained with hsp90 and hsp70 (Fig. 1C). Although gp96 was the most potent inducer of the four cytokines at comparable protein quantities, it is the least abundant among the HSPs tested. Hsp90 appears to be the most significant stimulator on a per cell equivalent basis when one considers that hsp90 is the most abundant among the HSPs. In addition, the ability of non-HSPs such as histone, ovalbumin and insulin was also tested in the same buffers as the HSPs, gp96, hsp90 or hsp70. No stimulation of cytokine release was elicited by these non-HSP's (Fig. 1D).

6.4 ABILITY TO ACTIVATE CD1 lb+ CELLS DOES NOT DERIVE FROM LPS As LPS is a known and potent stimulator of APCs and as LPS may contaminate buffers, the possibility that contaminating LPS may be responsible for the observed effects was tested, even though the HSP preparations used in the experiments shown in Fig. 1 were isolated from a mammalian source under clean conditions. Mice of the C3H/HeJ strain are known to be hypo-responsiveness to LPS and the ability of CD1 lb+ cells from these mice and their LPS-responsive counterparts, the C3H/HeN mice, was tested. It was observed that similar to LPS, gp96 preparations failed to stimulate CD1 lb+ cells from the C3H/HeJ strain to secrete TNF-ol or IL-lß (Fig. 2). At first look, these observations suggested that the APC-activating activity of HSP preparations was derived from contaminating LPS.

In order to explore the contribution of LPS more rigorously, HSPs were purified by the deliberate use of LPS-free reagents and Good Manufacturing Practices of the US Food and Drug Administration and the resulting HSP preparations were tested and shown to be free of detectable levels of LPS (<0.02 e. u. ) by Limulus amebocyte lysate (LAL) assay. The LPS-free HSP preparations still stimulated CD1 lb+ cells to secrete

cytokines as shown in Fig. 3A. As the lowest detection limit of LPS in the LAL assay in <BR> <BR> our hands was 0.02 e. u. , this quantity of LPS and ten times higher quantities of LPS were tested for their ability to activate APCs. These quantities were found to be too low to stimulate the APCs to release either of the two cytokines tested (Fig. 3A). Far larger quantities of LPS, i. e. 1000 e. u. /ml were necessary to stimulate the CD1 lb+ cells under the conditions used in our experiments (Fig. 3A). This is not a large quantity of LPS in itself, and is comparable to the quantities used in previous studies for activation of APCs. As additional control, under conditions where increasing quantities of gp96 induced the release of increasing levels of TNF-a and IL-ß (i. e. under linear conditions of the assay), twice the highest quantity of serum albumin prepared in the same buffer as gp96, did not lead to release of detectable levels of either cytokine (Fig. 3A).

The effect of LPS was tested in another manner. Activation of APCs by LPS is dependent upon the presence of the LPS-binding protein (LBP) normally present in serum. The LBP concentrates LPS and delivers it to CD14 molecules on the APC surface thus permitting relatively low concentrations of LPS to activate APCs. The LBP- dependence is less pronounced or absent at high LPS concentrations. In order to distinguish the roles of LPS and HSPs in APC-activation, serum dependence of each activity was tested. The adherent fraction of the PECs (over 90% CD1 lb+) of C57BL/6 mice was simulated with titrated quantities of gp96 or LPS preparations in the presence or absence of serum and the supernatants were tested for the presence of IL- (3. It was observed (Fig. 3B) that while LPS preparations were significantly dependent on the presence of serum, the ability of gp96 preparations to stimulate APCs to secrete IL-j6 was entirely unaffected by serum. These considerations ruled out the possibility that the observed activation of CD1 lb+ cells by HSP preparations is not due to LPS contaminants in the preparation but is inherent in the HSPs themselves.

The effect of LPS was tested in yet another manner by using an antagonist (competitive inhibitor) of LPS (Rslp), derived from Rhodopseudomonas spheroids <BR> <BR> (Henricson et al. , 1992, Infect Immun., 60: 4285-90). This inhibitor diminished the ability of LPS but not gp96 to stimulate secretion of IL-1, by >75%. In fact, the activity of gp96 was greater in the presence of Rslp (Fig. 3C).

6.5 HSPs STIMULATE EXPRESSION OF ANTIGEN PRESENTING AND COSTIMULATORY MOLECULES

The effect of HSPs on maturation of dendritic cells (DCs) was examined.

Homogenous, LPS-free preparations of the HSPs gp96 and hsp70 were obtained from livers of C57BL/6 mice. Bone marrow-derived DCs, obtained from culturing in GM-CSF- containing medium, were pulsed with gp96, hsp70, or LPS (as a positive control) or serum albumin (as a negative control). The pulsed DCs were tested for surface expression of MHC II, B7.1, B7.2 and CD40 molecules. LPS induced expression of all markers tested.

Gp96 (400, ug/ml) was observed to induce a high degree of expression of MHC II and the co-stimulatory molecule B7.2, but not B7.1 nor CD40 (Fig. 4). Hsp70 (400 jug/ml), on the other hand, elicited a modest stimulation of surface expression of B7.2 but not B7.1, nor MHC II and CD40. The complete lack of stimulation of CD40 expression by gp96 or hsp70 led us to test this phenomenon more extensively and at a range of concentrations of the HSPs (40-400, ug/ml) ; however, CD40 expression was not induced at any concentration tested. Serum albumin (400 ug/ml), in the same buffer as the HSPs, did not induce expression of any of the markers tested.

6.6. HSP ACTIVATES TRANSLOCATION OF NF-KB The mechanism through which gp96 interacts with APCs was investigated, with reference to the activation of the NFKB pathway, shown previously (Ghosh et al., 1998, Ann. Rev. Immunol. 16: 225-260) to be a key transcriptional regulator for several cytokines and other immunologically important molecules. This pathway has also been shown to be activated in response to LPS and to be involved in the maturation of dendritic cells (Rescigno et al., 1998, J. Exp. Med. 188 : 2175-2180). Primary cultures of CDl lc+ cells were pulsed with gp96 or LPS and cells were harvested at various time intervals.

Nuclear extracts from the samples were used for binding to NFKB-specific oligomers and were resolved by native PAGE. It was observed the gp96 activates the transduction pathway and does so with a kinetics distinctly different from that of LPS (Fig. 5). The nuclear translocation of NFKB is seen in gp96-treated dendritic cells (DCs) as early as 15 minutes after pulsing and the signal diminishes to background levels by 120 minutes. In contrast, the translocation in LPS-treated DCs has a slower initiation kinetics. The differences in the kinetics of translocation of NFKB between gp96 and LPS as seen here is not a function of the quantities of either agent. Exposure of DCs to graded quantities of each shows the same differences in kinetics. In addition to providing a key glimpse into the mechanism through which HSPs activate APCs, these studies show the extent to which the effects of LPS and HSPs on APCs are similar yet distinct.

In view of the data shown in Fig. 5, and in view of the recent demonstration by Gallucci et al. (1999, Nat. Med. 5: 1249-55) and Sauter et al. (2000, J. Exp. Med.

191: 423-434) the necrotic but not apoptotic cells mediate maturation of DCs, we tested whether exposure of DCs to necrotic or apoptotic cells leads to translocation of NFKB to the nucleus. Cultures of immature DCs were exposed to necrotic or apoptotic E. G7 cells (prepared as described in Methods), and were monitored for expression of MHC II, B7.1, B7.2 and CD40. Exposure of DCs to necrotic but not apoptotic cells elicited expression of several maturation markers on the DCs (Fig. 6A), and also elicited translocation of NFKB to the nucleus (Fig. 6B).

6. 7. DISCUSSION HSPs are intracellular molecules and the physiological relevance of their ability to activate APCs may not be immediately obvious. However, being the most abundant, soluble, intracellular molecules, the presence of HSPs in the extracellular milieu would act an excellent message alerting the APCs to physical damage of the surrounding cells, whether as a consequence of bacterial and viral infections or mechanical injury. The ability of this signal to activate APCs can therefore be easily considered to confer an immunological, and hence survival advantage to the organism. The co-segregation of immunogenicity of a variety of cancers with higher levels of expression of inducible hsp70, without any preceding change in the antigenic repertoire of the cancers, is a case in point <BR> <BR> (1995, Menoret et al. , J. Immunol., 155: 740-7; 1998, Melcher et al. , Nat. Med. , 5: 581-7).

Conversely, the lack of such a signal may provide a mechanism for discrimination between the presence of antigen with and without'danger', as proposed in Fuchs and Matziner (1996, Semin. Immunol., 8 : 271-80). The quantities of HSPs shown here to be necessary to stimulate APCs in vitro are well within the range expected to be released locally as a result of cell lysis in vivo. Typically, 1 g of tissue yields approximately 30, ug gp96, 200/. tg hsp70 and 400, ug hsp90. These recoveries are somewhere in the range of 25%. Thus, 1 g of tissue contains-2. 5 mg HSPs. Considering that the tissue lysis in vivo can be reasonably assumed to happen not in solution but in a semi-liquid physical state, lysis of as little as 1 mg of cells (approximately 105-106 cells, depending on the cell type) will lead to release of - 2jug HSP in a volume of ~1-2, u1 or less. That is a concentration of 1-2 mg/ml-a higher concentration than that used in the described studies in vitro. Considerations of quantity are therefore compatible with a role in vivo, of HSPs in activation of APCs.

Examination of the levels of cytokines released by DCs, or of the extent of induction of the maturation markers on DCs by stimulation with HSPs, shows that the HSPs stimulate the DCs to a modest degree, as compared with the stimulation conferred by LPS.

For this reason, the inventors have tested the observations repeatedly in as many as ten experiments and have found them to be consistent. The inventors infer that the endogenous activators of DCs (HSPs in this instance) are much slower activators than external activators such as LPS for a physiological reason: the lower'specific activity'of endogenous signals allows for a more regulated activity, as the response to an internal signal might have to be far more modulated and more titratable, than that to an external signal.

All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only, and the invention is to be limited only by the terms of the appended claims along with the full scope of equivalents to which such claims are entitled.