Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Virology ; 249(1): 89-97, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9740780

ABSTRACT

Using reverse genetics methods, we constructed three different transfectant influenza A viruses encoding an Ld-restricted, nine amino-acid-long fragment, corresponding to amino-acid residues 876-884, of beta-galactosidase (beta-gal). Sequences encoding this epitope were nested within the hemagglutinin (HA) or neuraminidase (NA) open reading frames. Alternatively, an independent beta-gal mini-gene, preceded by an endoplasmic reticulum insertion signal sequence, was placed in a bicistronic arrangement in the NA RNA segment of the virus. All three transfectants mediated the presentation of the epitope to a beta-gal-specific CTL clone. Furthermore, each of the three transfectant viruses expressing the beta-gal fragment elicited specific cytolytic responses in vivo. Most importantly, these H1N1 transfectants mediated the regression of established murine pulmonary metastases. Tumor regression in mice was also achieved in the presence of preexisting immunity against an H3N2 influenza A virus serotype. Nononcogenic and nonintegrating, transfectant influenza A viruses are attractive candidates for development as antitumor vaccines.


Subject(s)
Cytotoxicity, Immunologic , Influenza A virus/immunology , Neoplasms, Experimental/immunology , RNA/immunology , Amino Acid Sequence , Animals , Antigen Presentation , Epitopes , Female , Influenza A virus/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA/genetics
7.
J Immunol Methods ; 214(1-2): 51-62, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9692858

ABSTRACT

Lymphocytes from patients with melanoma have been used to clone melanoma associated antigens which are, for the most part, nonmutated melanocyte tissue differentiation antigens. To establish a mouse model for the use of these 'self' antigens as targets for anti-tumor immune responses, we have employed the mouse homologues of the human melanoma antigens Tyrosinase, Tyrosinase Related Protein-1 (TRP-1), gp100, and MART-1. We sought to generate antisera against these proteins for use in the construction of experimental recombinant and synthetic anti-cancer vaccines, and for use in biologic studies. Using genes cloned from the B16 mouse melanoma or from murine melanocytes, we immunized rabbits with plasmid DNAs coated onto microscopic gold beads that were then delivered using a hand-held, helium-driven 'gene gun'. This strategy enabled us to generate polyclonal rabbit sera containing antibodies that specifically recognized each antigen, as measured by immunostaining of vaccinia virus infected cells. The sera that we generated specifically for TRP-1, gp100, and MART-1 recognized extracts of the spontaneous murine melanoma, B16. The identities of the recognized proteins was confirmed by Western blot analysis. The titers and specificities of these antisera were determined using ELISA. Interestingly, serum samples generated against murine MART-1 and gp100 developed antibodies that were cross-reactive with the corresponding human homologues. Recognition of human gp100 and murine Tyrosinase appeared to be dependent upon conformational epitopes since specificity was lost upon denaturation of the antigens. These antisera may be useful in the detection, purification and characterization of the mouse homologues of recently cloned human tumor associated antigens and may enable the establishment of an animal model of the immune consequences of vaccination against 'self antigens.


Subject(s)
DNA, Neoplasm/administration & dosage , Genetic Therapy/methods , Immune Sera/biosynthesis , Immune Sera/immunology , Immunization, Passive/methods , Neoplasm Proteins/immunology , Animals , Antibody Specificity , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Blotting, Western , Cells, Cultured , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Enzyme-Linked Immunosorbent Assay , Haplorhini , Humans , Immune Sera/genetics , Kidney/virology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma-Specific Antigens , Mice , Neoplasm Proteins/genetics , Plasmids , Rabbits , Vaccinia virus/genetics , Vaccinia virus/metabolism
8.
J Natl Cancer Inst ; 89(21): 1595-601, 1997 Nov 05.
Article in English | MEDLINE | ID: mdl-9362157

ABSTRACT

BACKGROUND: The identification of tumor-associated antigens and the cloning of DNA sequences encoding them have enabled the development of anticancer vaccines. Such vaccines target tumors by stimulating an immune response against the antigens. One method of vaccination involves the delivery of antigen-encoding DNA sequences, and a number of recombinant vectors have been used for this purpose. To optimize the efficacy of recombinant vaccines, we compared primary and booster treatment regimens that used a single vector (i.e., homologous boosting) with regimens that used two different vectors (i.e., heterologous boosting). METHODS: Pulmonary tumors (experimental metastases) were induced in BALB/c mice inoculated with CT26.CL25 murine colon carcinoma cells, which express recombinant bacterial beta-galactosidase (the model antigen). Protocols for subsequent vaccination used three vectors that encoded beta-galactosidase--vaccinia (cowpox) virus, fowlpox virus, naked bacterial plasmid DNA. Mouse survival was evaluated in conjunction with antibody and cytotoxic T-lymphocyte responses to beta-galactosidase. RESULTS: Heterologous boosting resulted in significantly longer mouse survival than homologous boosting (all P<.0001, two-sided). Potent antigen-specific cytotoxic T lymphocytes were generated following heterologous boosting with poxvirus vectors. This response was not observed with any of the homologous boosting regimens. Mice primed with recombinant poxvirus vectors generated highly specific antibodies against viral proteins. CONCLUSIONS: The poor efficacy of homologous boosting regimens with viral vectors was probably a consequence of the induction of a strong antiviral antibody response. Heterologous boosting augmented antitumor immunity by generating a strong antigen-specific cytotoxic T-lymphocyte response. These data suggest that heterologous boosting strategies may be useful in increasing the efficacy of recombinant DNA anticancer vaccines that have now entered clinical trials.


Subject(s)
Cancer Vaccines/administration & dosage , Genetic Vectors , Lung Neoplasms/prevention & control , Animals , Blotting, Western , Cancer Vaccines/therapeutic use , DNA, Bacterial , DNA, Viral , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunization Schedule , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Survival Analysis
9.
Cell Growth Differ ; 8(7): 811-20, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9218875

ABSTRACT

Control of cell proliferation is dependent on the regulated expression of the cyclin genes. Induction of cyclin B1 gene expression in S phase has been shown to require sequences within the first 90 bp of the proximal promoter region. In this study, we defined the cell cycle regulatory elements within this region and explored the mechanism by which the cyclin B1 gene is activated. A CDE-like element that is important in S-phase regulation of other genes was not required for correct cell cycle expression of cyclin B1. Instead, two CCAAT boxes were essential for S-phase induction of cyclin B1 gene in both NIH3T3 and HeLa cells. Induction of cyclin B1 by cyclin/cyclin-dependent kinase (cdk) complexes were examined by cotransfection of the reporter along with appropriate expression vectors. Complexes of cdk4 with cyclin D1 or cdk2 with cyclin E or A can activate the cyclin B1 promoter, and activation is uniquely dependent on the CCAAT elements in both normal and heterologous contexts. This transcription factor NF-Y binds to both CCAAT elements. These findings suggest that S phase-specific induction of the cyclin B1 promoter is dependent upon NF-Y binding to the CCAAT elements and is correlated with activation by cyclin-dependent kinases.


Subject(s)
CDC2-CDC28 Kinases , Cyclin B , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins , S Phase/genetics , Transcriptional Activation/genetics , 3T3 Cells , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Cyclin B1 , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/genetics , Cyclins/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Mice , Molecular Sequence Data , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...