Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Vaccine ; 29(18): 3476-82, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21382485

ABSTRACT

Our laboratory previously described the strategy of fusing chemokine receptor ligands to antigens in order to generate immunogenic DNA vaccines. In the present study, we produced mouse ß-2 defensin (mBD2) fusion proteins using both ovalbumin (OVA) and gp100 as model antigens. Superior cross-presentation by dendritic cells (DC) was observed for mBD2 fused antigens over unfused antigens in vitro. In vivo, we observed significant increases in the expansion of adoptively transferred antigen-specific MHC class I, but not class II-restricted T cells after immunization with mBD2 fused antigen over antigen alone. This enhanced expansion of class I restricted T cells was Toll-like receptor 4 (TLR4) dependent, but CC chemokine receptor 6 (CCR6) independent. Superior tumor resistance was observed for mBD2-fusion protein vaccines, compared to unfused antigen, in both B16-OVA and B16 tumor models. These data suggest that production of mBD2 fusion proteins is feasible and that the vaccines facilitate in vivo expansion of adoptively transferred T cells through a TLR4-dependent mechanism.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Toll-Like Receptor 4/immunology , beta-Defensins/immunology , Animals , Antigen-Presenting Cells , Cross-Priming , Interferon-gamma/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Receptors, CCR6/immunology , Recombinant Fusion Proteins/immunology , gp100 Melanoma Antigen/immunology
2.
Cancer Res ; 61(21): 7920-4, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691813

ABSTRACT

Endogenous retroviral gene products have been found in some human tumors, and therefore, may serve as antigens for immunotherapy approaches. The murine colorectal carcinoma CT26 and melanoma B16 have recently been found to express the endogenous retroviral gene products gp70 and p15E, respectively, that can serve as antigens recognized by T cells. To date, though, there has been no demonstration of tumor treatment using an endogenous retroviral protein. In this study, we demonstrate that mice immunized with recombinant vaccinia encoding the gp70 H2-L(d)-restricted minimal determinant were protected from CT26 tumor challenge. Splenocytes from mice immunized with vaccinia gp70 specifically secreted IFN-gamma in response to gp70 peptide-pulsed stimulators. Although this strategy could protect against subsequent tumor challenge, it was ineffective against established tumors. Therefore, to investigate the treatment of established CT26 or B16 lung metastases, mice were treated with cultured dendritic cells (DCs) pulsed with gp70 or p15E peptide. Significant inhibition of established lung metastases required immunization with peptide-pulsed DCs pretreated with CD40 ligand that has been demonstrated to increase the T-cell stimulatory activity of DCs. The ability to immunize against endogenous retroviral tumor antigens may have relevance in the induction of antitumor immunity for some human cancers.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Cancer Vaccines/immunology , Retroviridae Proteins, Oncogenic/immunology , Viral Envelope Proteins/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , CD40 Ligand/immunology , Cancer Vaccines/genetics , Colonic Neoplasms/therapy , Dendritic Cells/immunology , Immunotherapy, Adoptive , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Retroviridae Proteins, Oncogenic/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Envelope Proteins/genetics
3.
J Exp Med ; 194(4): 481-9, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11514604

ABSTRACT

We have previously shown that small B16 melanomas can be successfully treated using a combination of anti-cytotoxic T lymphocyte antigen (CTLA)-4 monoclonal antibody with a granulocyte/macrophage colony-stimulating factor (GM-CSF) producing irradiated tumor cell vaccine. Regression of tumors results in long-lasting immunity and is frequently accompanied by autoimmune depigmentation. Here we examine the cellular and molecular mechanisms of this combined treatment. Histological examination of depigmented lesions revealed infiltration of polymorphonuclear cells and deposition of antibody. The combination therapy also induced tumor rejection and skin depigmentation in B cell-deficient and in CD4(+) T cell-depleted mice. Both effects of the treatment absolutely required CD8(+) T cells. Analysis of the response in successfully treated mice revealed elevated levels of CD8(+) T cells specific for a nonameric peptide consisting of residues 180-188 of the melanocyte differentiation antigen tyrosinase-related protein (TRP)2. There was no evidence of reactivity to the melanocyte antigens gp100, tyrosinase, Mart1/MelanA, or TRP1. Fas-FasL interactions and perforin played a role in mounting the effector response, whereas the tumor necrosis factor pathway was not required. The cellular requirements for tumor rejection in this therapeutic setting were strikingly different from those in a prophylactic setting. In particular, if mice received a prophylactic vaccine consisting of anti-CTLA-4 and B16-GM-CSF before tumor challenge, full protection was obtained even in the absence of CD8(+) T cells. Our data demonstrate that therapeutic autoreactive CD8(+) T cell responses can effectively be generated in tumor-bearing mice and stresses the value of studying tumor immunity in a therapeutic rather than a prophylactic setting.


Subject(s)
Antigens, Differentiation/immunology , Cancer Vaccines/immunology , Immunoconjugates , Melanoma, Experimental/therapy , Abatacept , Animals , Antigens, CD , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Cancer Vaccines/therapeutic use , Female , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL
4.
Trends Immunol ; 22(1): 5-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11286676

ABSTRACT

With the identification of tumor antigens and a knowledge of how to vaccinate against them, the field of tumor immunology faces new challenges. In this article, the authors argue that successful immunotherapies of the future will activate anti-tumor T cells without inducing their anergy or apoptotic death.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Active/trends , Animals , Humans , Immune Tolerance/immunology , Mice , Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Escape/immunology
5.
Blood ; 97(8): 2374-80, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290600

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) is associated with KS, primary effusion lymphoma (PEL), and multicentric Castleman disease. Reactivation of KSHV in latently infected cells and subsequent plasma viremia occur before the development of KS. Intracellular signaling pathways involved in KSHV reactivation were studied. In latently infected PEL cells (BCBL-1), KSHV reactivation in single cells was determined by quantitative flow cytometry. Viral particle production was determined by electron microscope analyses and detection of minor capsid protein in culture supernatants. Agents that mobilized intracellular calcium (ionomycin, thapsigargin) induced expression of KSHV lytic cycle-associated proteins and led to increased virus production. Calcium-mediated virus reactivation was blocked by specific inhibitors of calcineurin-dependent signal transduction (cyclosporine, FK506). Similarly, calcium-mediated virus reactivation in KSHV-infected dermal microvascular endothelial cells was blocked by cyclosporine. Furthermore, retroviral transduction with plasmid DNA encoding VIVIT, a peptide specifically blocking calcineurin-NFAT interactions, inhibited calcium-dependent KSHV reactivation. By contrast, chemical induction of lytic-phase infection by the phorbol ester 12-O-tetradecanoyl-phorbol-13-acetate was blocked by protein kinase C inhibitors, but not by calcineurin inhibitors. In summary, calcineurin-dependent signal transduction, an important signaling cascade in vivo, induces calcium-dependent KSHV replication, providing a possible target for the design of antiherpesvirus strategies in KSHV-infected patients.


Subject(s)
Calcineurin Inhibitors , Calcium Signaling/drug effects , Herpesvirus 8, Human/growth & development , Nuclear Proteins , Virus Activation/drug effects , Calcineurin/physiology , Calcium Signaling/physiology , Capsid/analysis , Cells, Cultured , Cyclosporine/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/virology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Indoles/pharmacology , Ionomycin/pharmacology , Maleimides/pharmacology , Microscopy, Electron , NFATC Transcription Factors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Protein Processing, Post-Translational , Staurosporine/pharmacology , Tacrolimus/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/drug effects , Transfection , Viral Proteins/biosynthesis
6.
Curr Protoc Immunol ; Chapter 20: Unit 20.1, 2001 May.
Article in English | MEDLINE | ID: mdl-18432774

ABSTRACT

This unit details protocols for in vivo models of subcutaneous growth and pulmonary metastases of B16 melanoma. Therapeutic approaches include the use of B16.GM-CSF and rVVmTRP-1 to induce autoimmune vitiligo and tumor protection. The induction and use of gp 100-specific therapeutic cytotoxic T lymphocytes (CTL) are discussed. Methods are also included for CTL induction, isolation and testing, CTL maintenance, and adoptive transfer. Support protocols detail the testing of mouse sera for presence of MDA-specific antibodies by immunoblotting and ELISA, respectively. Additional sections, including growing B16 melanoma, enumerating pulmonary metastases, and use of recombinant viruses for vaccination, are discussed together with safety concerns.


Subject(s)
Disease Models, Animal , Melanoma, Experimental , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunotherapy , Lung Neoplasms/secondary , Melanocytes/cytology , Melanocytes/enzymology , Melanocytes/immunology , Melanocytes/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Melanoma, Experimental/therapy , Mice , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/virology
7.
J Interferon Cytokine Res ; 20(6): 589-96, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10888115

ABSTRACT

Interleukin-12 (IL-12) has been shown to play a central role in the innate and acquired immune responses. Its activities include enhancement of natural killer (NK) and cytotoxic T lymphocyte (CTL) activity and promotion of CD4 Th1 cell development. It has also been shown to provide potent activity as a vaccine adjuvant in generating antibody and T cell responses. We have investigated the efficacy of IL-12 protein in promoting CD8 T cell responses when it is used as an adjuvant for immunization. Studies using, as antigen, cDNA from an autologous antigen (P1A) as well as studies of responses to vaccinia virus-delivered self (gp100) and non-self (beta-galactosidase) antigens show that the dose and schedule of IL-12 administration can significantly affect adjuvant activity, leading to enhancement or suppression of antigen-specific responses.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Interleukin-12/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , DNA, Complementary/genetics , DNA, Complementary/immunology , Dose-Response Relationship, Drug , Drug Administration Schedule , Immunization , Mast-Cell Sarcoma/drug therapy , Mast-Cell Sarcoma/immunology , Mast-Cell Sarcoma/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
8.
Proc Natl Acad Sci U S A ; 97(10): 5492-7, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10779556

ABSTRACT

We demonstrated that peripheral T cell tolerance toward murine melanoma self-antigens gp100 and TRP-2 can be broken by an autologous oral DNA vaccine containing the murine ubiquitin gene fused to minigenes encoding peptide epitopes gp100(25-33) and TRP-2(181-188). These epitopes contain dominant anchor residues for MHC class I antigen alleles H-2D(b) and H-2K(b), respectively. The DNA vaccine was delivered by oral gavage by using an attenuated strain of Salmonella typhimurium as carrier. Tumor-protective immunity was mediated by MHC class I antigen-restricted CD8(+) T cells that secreted T(H)1 cytokine IFN-gamma and induced tumor rejection and growth suppression after a lethal challenge with B16G3. 26 murine melanoma cells. Importantly, the protective immunity induced by this autologous DNA vaccine against murine melanoma cells was at least equal to that achieved through xenoimmunization with the human gp100(25-33) peptide, which differs in its three NH(2)-terminal amino acid residues from its murine counterpart and was previously reported to be clearly superior to an autologous vaccine in inducing protective immunity. The presence of ubiquitin upstream of the minigene proved to be essential for achieving this tumor-protective immunity, suggesting that effective antigen processing and presentation may make it possible to break peripheral T cell tolerance to a self-antigen. This vaccine design might prove useful for future rational designs of other recombinant DNA vaccines targeting tissue differentiation antigens expressed by tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Melanoma, Experimental/immunology , Vaccines, DNA/therapeutic use , Administration, Oral , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Female , Histocompatibility Antigens Class I/immunology , Humans , Male , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, SCID , Prostatic Neoplasms/immunology , Salmonella typhimurium/immunology , Ubiquitins/genetics , Vaccines, DNA/administration & dosage
9.
J Exp Med ; 191(7): 1221-32, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10748239

ABSTRACT

The human tyrosinase-derived peptide YMDGTMSQV is presented on the surface of human histocompatibility leukocyte antigen (HLA)-A*0201(+) melanomas and has been suggested to be a tumor antigen despite the fact that tyrosinase is also expressed in melanocytes. To gain information about immunoreactivity and self-tolerance to this antigen, we established a model using the murine tyrosinase-derived homologue of this peptide FMDGTMSQV, together with transgenic mice expressing the HLA-A*0201 recombinant molecule AAD. The murine peptide was processed and presented by AAD similarly to its human counterpart. After immunization with recombinant vaccinia virus encoding murine tyrosinase, we detected a robust AAD-restricted cytotoxic T lymphocyte (CTL) response to FMDGTMSQV in AAD transgenic mice in which the entire tyrosinase gene had been deleted by a radiation-induced mutation. A residual response was observed in the AAD(+)tyrosinase(+) mice after activation under certain conditions. At least some of these residual CTLs in AAD(+)tyrosinase(+) mice were of high avidity and induced vitiligo upon adoptive transfer into AAD(+)tyrosinase(+) hosts. Collectively, these data suggest that FMDGTMSQV is naturally processed and presented in vivo, and that this presentation leads to substantial but incomplete self-tolerance. The relevance of this model to an understanding of the human immune response to tyrosinase is discussed.


Subject(s)
Antigens, Neoplasm/immunology , HLA-A2 Antigen/immunology , Melanoma/immunology , Monophenol Monooxygenase/immunology , Self Tolerance/immunology , Amino Acid Sequence , Animals , Antigen Presentation , Cross Reactions , HLA-A2 Antigen/genetics , Humans , Immunotherapy , Melanocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Monophenol Monooxygenase/genetics , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology
10.
Cancer Res ; 60(2): 253-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667570

ABSTRACT

"Self" melanocyte differentiation antigens are potential targets for specific melanoma immunotherapy. Vaccination against murine tyrosinase-related protein (TRP)-1/gp75 was shown recently to cause melanoma rejection, which was accompanied by autoimmune skin depigmentation (vitiligo). To further explore the linkage between immunotherapy and autoimmunity, we studied the response to vaccination with a related antigen, TRP-2. i.m. inoculation of plasmid DNA encoding murine trp-2 elicited antigen-specific CTLs that recognized the B16 mouse melanoma and protected the mice from challenge with tumor cells. Furthermore, mice bearing established s.c. B16 melanomas rejected the tumor upon vaccination with a recombinant vaccinia virus encoding trp-2. Depletion experiments showed that CD8+ lymphocytes and natural killer cells were crucial for the antitumor activity of the trp-2-encoding vaccines. Mice that rejected the tumor did not develop generalized vitiligo, indicating that protective immunity can be achieved in the absence of widespread autoimmune aggression.


Subject(s)
Cancer Vaccines/therapeutic use , Interferon Type I/immunology , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Pregnancy Proteins/immunology , Vaccines, Synthetic/therapeutic use , Vitiligo/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/toxicity , Killer Cells, Natural/immunology , Lymphocyte Culture Test, Mixed , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/chemistry , Peptides/immunology , Vaccines, Synthetic/toxicity , Vitiligo/etiology
11.
Cancer J Sci Am ; 6 Suppl 1: S76-80, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685664

ABSTRACT

PURPOSE: The purpose of our efforts is to trigger the immune destruction of established cancer. Interleukin (IL)-2 can mediate the regression of tumors in patients with melanoma and renal cell carcinoma. In animal models, the antitumor effects of IL-2 are mediated by T lymphocytes. Stimulation with specific antigen can enhance the ability of T cells to respond to IL-2 by triggering the rapid upregulation of the high-affinity IL-2 receptor. We are seeking to design recombinant and synthetic vaccines capable of preferentially priming T cells with specificity for tumor cells. METHODS: The antitumor activity of experimental vaccines is being studied preclinically using recently developed murine models that employ the mouse homologues of human tumor-associated antigens. Once the most effective experimental vaccines are optimized in experimental animals, clinical trials can be conducted. Vaccines are being evaluated for their ability to mediate the regression of established tumors, and a variety of immunologic correlates are being measured. RESULTS: In animal models, vaccines based on molecularly defined tumor-associated antigens expressed in viral vectors or delivered as "naked" DNA stimulate the expansion of CD4+ and CD8+ tumor-specific T lymphocytes. Coadministration of IL-2 with these vaccines dramatically enhances their ability to mediate the regression of established cancer. In the clinic, treatment of melanoma patients with peptide vaccine and IL-2 resulted in objective responses in approximately 40% of patients, a response rate more than twice that typically achieved with IL-2 alone. Paradoxically, tumor-specific CD8+ T-cell levels were not increased in these patients. CONCLUSION: The addition of recombinant and synthetic cancer vaccines to a regimen of IL-2 can result in improved antitumor responses in both animal models and melanoma patients. Vaccine-primed, tumor-specific T cells may preferentially proliferate upon administration of IL-2. The apparent lack of increase in CD8+ T-cell numbers in this setting suggests that the vaccine-primed T cells functionally disappear after a transient period of activation. Preventing the disappearance of activated T cells upon IL-2 administration-for example, by blocking proapoptotic signals-may enhance the therapeutic effectiveness of anticancer vaccines.


Subject(s)
Cancer Vaccines/immunology , Interleukin-2/therapeutic use , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Immunotherapy, Adoptive , Lymphocyte Activation , Mice , Neoplasms/immunology
12.
J Immunol ; 164(2): 562-5, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623795

ABSTRACT

Neoantigens resulting from the inherent genomic instability of tumor cells generally do not trigger immune recognition. Similarly, transfection of tumors with model Ags often fails to elicit CD8+ T cell responses or alter a tumor's growth rate or lethality. We report here that the adoptive transfer of activated Th1-type CD4+ T cells specific for a model tumor Ag results in the de novo generation of CD8+ T cells with specificity to that Ag and concomitant tumor destruction. The anti-tumor effects of the CD4+ T cells required the presence of both MHC class I and class II on host cells, as evidenced by experiments in knockout mice, suggesting that CD4+ T cells enhanced the ability of host APC to activate endogenous CD8+ T cells. These results indicate that the apparent inability of tumor cells expressing highly immunogenic epitopes to activate tumor-specific CD8+ T cells can be altered by activated CD4+ T cells.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , beta-Galactosidase/immunology , Adoptive Transfer , Animals , Cell Movement/immunology , Clone Cells/immunology , Clone Cells/transplantation , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/physiology , Histocompatibility Antigens Class II/physiology , Mice , Mice, Inbred C57BL , Models, Immunological , Neoplasm Transplantation , Sarcoma, Experimental/enzymology , Sarcoma, Experimental/immunology , Sarcoma, Experimental/therapy , Tumor Cells, Cultured , beta-Galactosidase/biosynthesis
13.
Crit Rev Immunol ; 20(6): 433-50, 2000.
Article in English | MEDLINE | ID: mdl-11396680

ABSTRACT

It is increasingly clear that immunity to "self"-antigens may result in tumor destruction in mouse and man. But which antigens should be targeted with therapeutic cancer vaccines? In the case of melanoma, recognition of melanocyte differentiation antigens (MDA) can be associated with autoimmune depigmentation (vitiligo). We propose that intersection of protein transport to melanosomes and endosomes allows for the loading of MDA-derived peptides on MHC class II molecules, resulting in the activation of MDA-specific CD4+ "helper" T cells that aid the induction of melanoma-specific CD8+ T cells. Thus, the immunogenicity of MDA may be a consequence of their unique cell biology. Studies of MDA-based vaccines can provide new insight into the development of more effective cancer vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Melanocytes/immunology , Melanoma/therapy , Membrane Glycoproteins/therapeutic use , Neoplasm Proteins/therapeutic use , Animals , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Male , Melanoma/immunology , Mice , Vitiligo/immunology , gp100 Melanoma Antigen
14.
Proc Natl Acad Sci U S A ; 96(6): 2982-7, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077623

ABSTRACT

Many human and mouse tumor antigens are normal, nonmutated tissue differentiation antigens. Consequently, immunization with these "self" antigens could induce autoimmunity. When we tried to induce immune responses to five mouse melanocyte differentiation antigens, gp100, MART-1, tyrosinase, and tyrosinase-related proteins (TRP) 1 and TRP-2, we observed striking depigmentation and melanocyte destruction only in the skin of mice inoculated with a vaccinia virus encoding mouse TRP-1. These mice rejected a lethal challenge of B16 melanoma, indicating the immune response against TRP-1 could destroy both normal and malignant melanocytes. Cytotoxic T lymphocytes specific for TRP-1 could not be detected in depigmented mice, but high titers of IgG anti-TRP-1 antibodies were present. Experiments with knockout mice revealed an absolute dependence on major histocompatibility complex class II, but not major histocompatibility complex class I, for the induction of both vitiligo and tumor protection. Together, these results suggest that the deliberate induction of self-reactivity using a recombinant viral vector can lead to tumor destruction, and that in this model, CD4(+) T lymphocytes are an integral part of this process. Vaccine strategies targeting tissue differentiation antigens may be valuable in cancers arising from nonessential cells and organs such as melanocytes, prostate, testis, breast, and ovary.


Subject(s)
Antigens, Neoplasm/immunology , Autoantigens/immunology , Cancer Vaccines , Cytotoxicity, Immunologic , Melanoma, Experimental/immunology , Membrane Glycoproteins , Oxidoreductases , Proteins/immunology , Vitiligo/immunology , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA, Recombinant , Female , Genetic Vectors , Humans , Male , Melanocytes/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Knockout , Vaccination , Vaccinia virus
15.
J Natl Cancer Inst ; 90(24): 1881-7, 1998 Dec 16.
Article in English | MEDLINE | ID: mdl-9862625

ABSTRACT

BACKGROUND: Construction of recombinant viruses that can serve as vaccines for the treatment of experimental murine tumors has recently been achieved. The cooperative effects of immune system modulators, including cytokines such as interleukin 12 (IL-12) and costimulatory molecules such as B7-1, may be necessary for activation of cytotoxic T lymphocytes. Thus, we have explored the feasibility and the efficacy of inclusion of these immunomodulatory molecules in recombinant virus vaccines in an experimental antitumor model in mice that uses Escherichia coli beta-galactosidase as a target antigen. METHODS: We developed a "cassette" system in which three loci of the vaccinia virus genome were used for homologous recombination. A variety of recombinant vaccinia viruses were constructed, including one virus, vB7/beta/IL-12, that contains the following five transgenes: murine B7-1, murine IL-12 subunit p35, murine IL-12 subunit p40, E. coli lacZ (encodes beta-galactosidase, the model antigen), and E. coli gpt (xanthine-guanine phosphoribosyltransferase, a selection gene). The effects of the recombinant viruses on lung metastases and survival were tested in animals that had been given an intravenous injection of beta-galactosidase-expressing murine colon carcinoma cells 3 days before they received the recombinant virus by intravenous inoculation. RESULTS: Expression of functional B7-1 and IL-12 by virally infected cells was demonstrated in vitro. Lung tumor nodules (i.e., metastases) were reduced in mice by more than 95% after treatment with the virus vB7/beta/IL-12; a further reduction in lung tumor nodules was observed when exogenous IL-12 was also given. Greatest survival of tumor-bearing mice was observed in those treated with viruses encoding beta-galactosidase and B7-1 plus exogenous IL-12. CONCLUSION: This study shows the feasibility of constructing vaccinia viruses that express tumor antigens and multiple immune cofactors to create unique immunologic microenvironments that can modulate immune responses to cancer.


Subject(s)
Antigens, Neoplasm/genetics , B7-1 Antigen/genetics , Cancer Vaccines/genetics , Interleukin-12/genetics , Neoplasms, Experimental/therapy , Vaccinia virus/genetics , Animals , B7-1 Antigen/immunology , Blotting, Western , Cancer Vaccines/therapeutic use , Escherichia coli/enzymology , Escherichia coli/genetics , Feasibility Studies , Female , Flow Cytometry , Interleukin-12/immunology , Lac Operon , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Pentosyltransferases/genetics , Tumor Cells, Cultured , Vaccinia virus/immunology , beta-Galactosidase/genetics , beta-Galactosidase/immunology
16.
J Immunol ; 161(10): 5313-20, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9820504

ABSTRACT

Following an infection or immunization, a primary CD8+ T cell response generally rises then falls rapidly before giving rise to a "memory" response. When we immunized mice with recombinant viral immunogens optimized to enhance the lytic capability of CD8+ T cells, we measured a profound depression in Ag-specific effector function after early restimulation. Indeed, a "mirror image" cytolytic capability was observed: the most powerful immunogens, as measured by cytolytic capacity 6 days after immunization, elicited the weakest secondary immune response when evaluated following an additional 6 days after restimulation. To understand the mechanism of this suppression, we examined the fate of splenocytes immunized with a vaccinia virus encoding Ag and IL-2 then restimulated ex vivo. We found that these splenocytes underwent an apoptotic cell death, upon early restimulation, that was not dependent on the engagement of the FasR (CD95). Unlike previously described mechanisms of "propriocidal cell death" and "clonal exhaustion," the cell death we observed was not an inherent property of the CD8+ T cells but rather was due to a population of splenocytes that stained positive for both the Mac-1 and Gr-1 surface markers. Deletion of these cells in vitro or in vivo completely abrogated the observed suppression of cytolytic reactivity of Ag-specific CD8+ T cells. These observations could account for the apparent absence of Ag-specific immune responses after some current vaccination regimens employing powerful immunogens. Finally, our results may shed new light on a mechanism for the suppression of CD8+ T cell responses and its effect on vaccine efficacy and on immune memory.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Macrophage-1 Antigen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Communication/immunology , Cell Membrane/immunology , Cytotoxicity, Immunologic , Female , HeLa Cells , Humans , Immune Tolerance , Immunization , Immunization, Secondary , Interleukin-2/genetics , Lymphocyte Count , Lymphocyte Depletion , Macrophage-1 Antigen/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Vaccinia virus/genetics , Vaccinia virus/immunology , beta-Galactosidase/genetics
17.
J Exp Med ; 188(2): 277-86, 1998 Jul 20.
Article in English | MEDLINE | ID: mdl-9670040

ABSTRACT

Many tumor-associated antigens are nonmutated, poorly immunogenic tissue differentiation antigens. Their weak immunogenicity may be due to "self"-tolerance. To induce autoreactive T cells, we studied immune responses to gp100/pmel 17, an antigen naturally expressed by both normal melanocytes and melanoma cells. Although a recombinant vaccinia virus (rVV) encoding the mouse homologue of gp100 was nonimmunogenic, immunization of normal C57BL/6 mice with the rVV encoding the human gp100 elicited a specific CD8(+) T cell response. These lymphocytes were cross-reactive with mgp100 in vitro and treated established B16 melanoma upon adoptive transfer. To understand the mechanism of the greater immunogenicity of the human version of gp100, we characterized a 9-amino acid (AA) epitope, restricted by H-2Db, that was recognized by the T cells. The ability to induce specific T cells with human but not mouse gp100 resulted from differences within the major histocompatibility complex (MHC) class I-restricted epitope and not from differences elsewhere in the molecule, as was evidenced by experiments in which mice were immunized with rVV containing minigenes encoding these epitopes. Although the human (hgp10025-33) and mouse (mgp10025-33) epitopes were homologous, differences in the three NH2-terminal AAs resulted in a 2-log increase in the ability of the human peptide to stabilize "empty" Db on RMA-S cells and a 3-log increase in its ability to trigger interferon gamma release by T cells. Thus, the fortuitous existence of a peptide homologue with significantly greater avidity for MHC class I resulted in the generation of self-reactive T cells. High-affinity, altered peptide ligands might be useful in the rational design of recombinant and synthetic vaccines that target tissue differentiation antigens expressed by tumors.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Proteins/immunology , Adoptive Transfer , Animals , Cytotoxicity, Immunologic , Female , Histocompatibility Antigens Class I/immunology , Humans , Ligands , Melanoma/immunology , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Tumor Escape , gp100 Melanoma Antigen
18.
Methods ; 12(2): 117-23, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9184376

ABSTRACT

The use of recombinant and synthetic vaccines in the treatment of cancer has recently been explored using model tumor associated antigens (TAA), many of which do not model the immunological state of affairs in which the TAA is expressed by normal tissues. One potentially useful model Ag is beta-galactosidase (beta-gal). Because the activity of this enzyme is so easily detectable, this gene has been inserted into a large number of recombinant viruses and tumors useful to the cancer vaccinologist. In addition, numerous transgenic mouse colonies that have tissue-specific expression of beta-gal have been developed, enabling the modeling of tolerance to "self" Ags. Since most of these mice have an H-2b background, we generated cytotoxic T lymphocytes (CTL) capable of recognizing beta-gal-expressing tumor cells of C57BL\6 origin and have determined that their restriction element is the K(b) molecule. Using an allele-specific epitope forecast to generate a panel of candidate peptides, we have determined that the K(b)-restricted sequence is DAPIYTNV and corresponds to amino acids 96-103 of the intact beta-gal molecule. A recombinant vaccinia virus (rVV-ES beta-gal96-103) was constructed that encoded the peptide epitope preceded by an endoplasmic reticulum insertion signal sequence. Tumor cells infected with this rVV were recognized by the original CTL that had been used to identify the epitope. Furthermore, splenocytes of mice immunized with a rVV encoding the full-length beta-gal molecule and restimulated with the DAPIYTNV peptide specifically recognized tumor cells expressing beta-gal. The identification of this immunogenic beta-gal sequence enables the modeling of immunization strategies in animal models of malignant disease in which the target antigen is a "self" protein.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Immunotherapy, Active/methods , Oligopeptides/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , beta-Galactosidase/immunology , Amino Acid Sequence , Animals , Cancer Vaccines/therapeutic use , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-gamma/analysis , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/therapy , Oligopeptides/chemistry , Vaccines, Synthetic/therapeutic use , Vaccinia virus/genetics , beta-Galactosidase/chemistry , beta-Galactosidase/genetics
19.
Proc Natl Acad Sci U S A ; 94(7): 3183-8, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9096367

ABSTRACT

Recombinant poxviruses encoding tumor-associated antigens (TAA) are attractive as candidate cancer vaccines. Their effectiveness, however, will depend upon expression of the TAA in appropriate antigen-presenting cells. We have used a murine model in which the TAA is beta-galactosidase (beta-gal) and a panel of recombinant vaccinia viruses (rVV) in which beta-gal was expressed under early or late promoters at levels that varied over 500-fold during productive infections in tissue culture cells. Remarkably, only those rVV employing early promoters were capable of prolonging the survival of mice bearing established tumors expressing the model TAA. Late promoters were ineffective regardless of their determined promoter strength. The best results were obtained when beta-gal was regulated by a strong early promoter coupled to a strong late promoter. When a variety of cell types were infected with the panel of viruses in vitro, dendritic cells were found to express beta-gal only under the control of the early promoters even though late promoters were intrinsically more active in other cell types. Furthermore, in a functional assay, dendritic cells infected in vitro with rVV encoding beta-gal regulated by an early promoter activated beta-gal-specific cytotoxic T lymphocytes, whereas similar rVV with a late promoter-regulated gene did not. These data indicate that promoter strength per se is not the most critical quality of a recombinant poxvirus-based tumor vaccine and that the use of promoters capable of driving the production of TAA in "professional" antigen presenting cells may be crucial.


Subject(s)
Antigens/biosynthesis , Cancer Vaccines/immunology , Dendritic Cells/immunology , Vaccines, Synthetic/immunology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Female , Humans , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Promoter Regions, Genetic , Vaccines, Synthetic/genetics , Vaccines, Synthetic/therapeutic use , Vaccinia virus/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
20.
Vaccine ; 15(4): 387-94, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9141209

ABSTRACT

Modified vaccinia virus Ankara (MVA), a highly attenuated strain of vaccinia virus (VV) that is unable to replicate in most mammalian cells, was evaluated as an expression vector for a model tumor associated antigen (TAA) and as a potential anti-cancer vaccine. We employed an experimental murine model in which an adenocarcinoma tumor line, CT26.CL25, was stably transfected with a model TAA, beta-galactosidase (beta-gal). Mice injected intramuscularly with a recombinant MVA (rMVA) expressing beta-gal (MVA-LZ), were protected from a lethal intravenous (i.v.) challenge with CT26.CL25. In addition, splenocytes from mice primed with MVA-LZ were therapeutically effective upon adoptive transfer to mice bearing pulmonary metastases of the CT26.CL25 tumor established 3 days earlier. Most importantly, i.v. inoculation with MVA-LZ resulted in significantly prolonged survival of mice bearing three day old pulmonary metastases. This prolonged survival compared favorably to mice treated with a replication competent recombinant VV expressing beta-gal. These findings indicate that rMVA is an efficacious alternative to the more commonly used replication competent VV for the development of new recombinant anti-cancer vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Genetic Vectors/immunology , Vaccinia virus/immunology , Adenocarcinoma/mortality , Adenocarcinoma/prevention & control , Adenocarcinoma/secondary , Animals , Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Antigens, Tumor-Associated, Carbohydrate/immunology , Colonic Neoplasms/immunology , Cytokines/biosynthesis , Disease Models, Animal , Female , Immunization, Secondary , Immunotherapy, Adoptive/methods , Lung Neoplasms/mortality , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Spleen/immunology , Spleen/metabolism , Spleen/transplantation , Tumor Cells, Cultured , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/physiology , Virus Replication , beta-Galactosidase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...