Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunother ; 22(6): 489-96, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10570747

ABSTRACT

Interleukin-10 (IL-10) has a wide range of in vivo biological activities and is a key regulatory cytokine of immune-mediated inflammation. The authors found that murine IL-10 given 12 hours after a recombinant vaccinia virus (rVV) containing the LacZ gene significantly enhanced the treatment of mice bearing 3-day-old pulmonary metastases expressing beta-galactosidase. Because IL-10 has been shown to inhibit the functions of key elements of both innate and acquired immune responses, the authors hypothesized that IL-10 might act by inhibiting clearance of the rVV, thus prolonging exposure to the experimental antigen. However, evidence that IL-10 was not acting primarily through such negative regulatory mechanisms included the following: (a) IL-10 also enhanced the therapeutic effectiveness of a recombinant fowlpox virus, which cannot replicate in mammalian cells; (b) Titers of rVV in immunized mice were lower, not higher; and (c) Although IL-10 did not alter levels of anti-vaccinia anti-bodies or natural killer cell activity, rVV-primed mice treated with IL-10 had enhanced vaccinia-specific cytotoxic T-lymphocyte activity. Thus, IL-10 enhanced the function of a recombinant poxvirus-based anti-cancer vaccine and may represent a potential adjuvant in the vaccination against human cancers using recombinant poxvirus-based vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Interleukin-10/administration & dosage , Neoplasms, Experimental/therapy , Vaccines, Synthetic/therapeutic use , Vaccinia virus/immunology , Adenocarcinoma/therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Colonic Neoplasms/therapy , Female , Humans , Interleukin-10/therapeutic use , Kinetics , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccinia virus/genetics , beta-Galactosidase/genetics , beta-Galactosidase/immunology
2.
J Natl Cancer Inst ; 91(20): 1744-50, 1999 Oct 20.
Article in English | MEDLINE | ID: mdl-10528025

ABSTRACT

BACKGROUND: Several approaches to gene therapy for cancer have yielded promising results in rodent models. The translation of these results to the clinical realm has been delayed by the lack of tumor models in large animals. We investigated the pattern of transgene (i. e., foreign or introduced gene) expression and virus vector elimination after systemic gene delivery using a thymidine kinase-negative vaccinia virus in a rabbit model of disseminated liver metastases. METHODS: VX-2 rabbit carcinoma cells were maintained by serial transplantation in the thigh muscles of New Zealand White rabbits, and disseminated liver metastases were established by direct injection of tumor cells into the portal vein of the animals. Different doses of a recombinant thymidine kinase-negative vaccinia virus vector encoding the firefly luciferase reporter gene (i.e., transgene) were injected into tumor-bearing rabbits. Transgene activity in tumors and other organs was measured at multiple time points thereafter. The pattern of development of antibodies against the vaccinia virus vector was also examined. Two-tailed Student's paired t test was used for comparisons of transgene activity. RESULTS: Transgene expression was increased in tumors by at least 16-fold in comparison with expression in other tissues by day 4 after vector injection (all P<. 001) and was maintained for approximately 1 week, providing evidence of tumor-specific gene delivery in this model. Rapid elimination of the circulating vector by the host immune system was observed. Anti-vector antibodies were detectable in serum as early as day 6 and were maintained for more than 3 months. CONCLUSIONS: Tumor-specific gene delivery is possible after systemic injection of a thymidine kinase-negative vaccinia virus vector in a model of rabbit liver metastases. Although the period of transgene expression appears limited because of a rapid immune response, the therapeutic window might be sufficient for an enzyme/prodrug gene therapy approach in clinical application.


Subject(s)
Gene Expression , Genetic Therapy/methods , Genetic Vectors , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/therapy , Transgenes/genetics , Vaccinia virus/genetics , Animals , Antibodies, Viral/blood , Cell Line , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Genes, Insect/genetics , Genes, Reporter/genetics , Genes, Viral/genetics , Genetic Vectors/therapeutic use , Haplorhini , HeLa Cells , Humans , Liver Neoplasms, Experimental/secondary , Luciferases/genetics , Plasmids , Rabbits , Recombinant Proteins/genetics , Thymidine Kinase/deficiency , Thymidine Kinase/genetics , Time Factors , Vaccinia virus/enzymology , Vaccinia virus/immunology
3.
Nat Med ; 5(7): 823-7, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10395329

ABSTRACT

'Naked' nucleic acid vaccines are potentially useful candidates for the treatment of patients with cancer, but their clinical efficacy has yet to be demonstrated. We sought to enhance the immunogenicity of a nucleic acid vaccine by making it 'self-replicating'. We accomplished this by using a gene encoding an RNA replicase polyprotein derived from the Semliki forest virus, in combination with a model antigen. A single intramuscular injection of a self-replicating RNA immunogen elicited antigen-specific antibody and CD8+ T-cell responses at doses as low as 0.1 microg. Pre-immunization with a self-replicating RNA vector protected mice from tumor challenge, and therapeutic immunization prolonged the survival of mice with established tumors. The self-replicating RNA vectors did not mediate the production of substantially more model antigen than a conventional DNA vaccine did in vitro. However, the enhanced efficacy in vivo correlated with a caspase-dependent apoptotic death in transfected cells. This death facilitated the uptake of apoptotic cells by dendritic cells, providing a potential mechanism for enhanced immunogenicity. Naked, non-infectious, self-replicating RNA may be an excellent candidate for the development of new cancer vaccines.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Colonic Neoplasms/prevention & control , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/therapeutic use , Animals , Antibody Formation , Apoptosis , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Enhancer Elements, Genetic , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Plasmids , Promoter Regions, Genetic , RNA-Dependent RNA Polymerase/biosynthesis , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Semliki forest virus/enzymology , Semliki forest virus/genetics , Transfection , Tumor Cells, Cultured
4.
Cancer Res ; 59(11): 2536-40, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10363968

ABSTRACT

To study the induction of anti-"self" CD8+ T-cell reactivity against the tumor antigen gp100, we used a mouse transgenic for a chimeric HLA-A*0201/H-2 Kb molecule (A2/Kb). We immunized the mice with a recombinant vaccinia virus encoding a form of gp100 that had been modified at position 210 (from a threonine to a methionine) to increase epitope binding to the restricting class I molecule. Immunogens containing the "anchor-fixed" modification elicited anti-self CD8+ T cells specific for the wild-type gp100(209-217) peptide pulsed onto target cells. More important, these cells specifically recognized the naturally presented epitope on the surface of an A2/Kb-expressing murine melanoma, B16. These data indicate that anchor-fixing epitopes could enhance the function of recombinant virus-based immunogens.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , HLA-A Antigens/immunology , Melanoma, Experimental/immunology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Transfection , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Autoimmunity/genetics , Cancer Vaccines/genetics , Epitopes/immunology , HLA-A Antigens/genetics , Immunity, Cellular , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Peptide Fragments/genetics , Vaccinia virus/genetics , Vaccinia virus/immunology , gp100 Melanoma Antigen
5.
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
6.
J Immunol ; 161(9): 4563-71, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9794383

ABSTRACT

CD40/CD40 ligand interactions have a central role in the induction of both humoral and cellular immunity. In this study, we examined whether a plasmid expressing CD40 ligand/trimer (CD40LT) could enhance immune responses in vivo. BALB/c mice were injected with plasmid expressing beta-galactosidase DNA with or without CD40LT DNA or IL-12 DNA, and immune responses were assessed. Mice vaccinated with beta-gal DNA plus CD40LT DNA or IL-12 DNA had a striking increase in Ag-specific production of IFN-gamma, cytolytic T cell activity, and IgG2a Ab. The mechanism by which CD40LT DNA enhanced these responses was further assessed by treating vaccinated mice with anti-IL-12 mAb or CTLA-4 Ig (CTLA4Ig). Production of IFN-gamma and CTL activity was abrogated by these treatments, suggesting that CD40LT DNA was mediating its effects on IFN-gamma and CTL activity through induction of IL-12 and enhancement of B7 expression, respectively. Physiologic relevance for the ability of CD40LT DNA to enhance immune responses by the aforementioned pathways was shown in two in vivo models. First, with regard to CTL activity, mice vaccinated with CD40LT DNA did not develop metastatic tumor following challenge with lethal dose of tumor. Moreover, in a mouse model requiring IL-12-dependent production of IFN-gamma, mice vaccinated with soluble Leishmania Ag and CD40LT DNA were able to control infection with Leishmania major. These data suggest that CD40LT DNA could be a useful vaccine adjuvant for diseases requiring cellular and/or humoral immunity.


Subject(s)
Adjuvants, Immunologic , Antibody Formation/drug effects , DNA, Recombinant/immunology , Immunity, Cellular/drug effects , Lung Neoplasms/secondary , Membrane Glycoproteins/genetics , Vaccines, Synthetic/immunology , Animals , Antibodies, Neoplasm/biosynthesis , B7-1 Antigen/immunology , CD28 Antigens/immunology , CD40 Ligand , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cytotoxicity, Immunologic , DNA, Recombinant/pharmacology , Female , Genes, Reporter , Immunoglobulin G/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-12/genetics , Interleukin-12/physiology , Leishmania major/immunology , Leishmaniasis/prevention & control , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Single-Blind Method , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Vaccination , beta-Galactosidase/genetics
7.
J Exp Med ; 188(6): 1075-82, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9743526

ABSTRACT

Cutaneous gene (DNA) bombardment results in substantial expression of the encoded antigen in the epidermal layer as well as detectable expression in dendritic cells (DC) in draining lymph nodes (LNs). Under these conditions, two possible modes of DC antigen presentation to naive CD8+ T cells might exist: (a) presentation directly by gene-transfected DC trafficking to local lymph nodes, and (b) cross-presentation by untransfected DC of antigen released from or associated with transfected epidermal cells. The relative contributions of these distinct modes of antigen presentation to priming for cytotoxic T cell (CTL) responses have not been clearly established. Here we show that LN cells directly expressing the DNA-encoded antigen are rare; 24 h after five abdominal skin bombardments, the number of these cells does not exceed 50-100 cells in an individual draining LN. However, over this same time period, the total number of CD11c+ DC increases more than twofold, by an average of 20,000-30,000 DC per major draining node. This augmentation is due to gold bombardment and is independent of the presence of plasmid DNA. Most antigen-bearing cells in the LNs draining the site of DNA delivery appear to be DC and can be depleted by antibodies to an intact surface protein encoded by cotransfected DNA. This finding of predominant antigen presentation by directly transfected cells is also consistent with data from studies on cotransfection with antigen and CD86-encoding DNA, showing that priming of anti-mutant influenza nucleoprotein CTLs with a single immunization is dependent upon coexpression of the DNAs encoding nucleoprotein and B7.2 in the same cells. These observations provide insight into the relative roles of direct gene expression and cross-presentation in CD8+ T cell priming using gene gun immunization, and indicate that augmentation of direct DC gene expression may enhance such priming.


Subject(s)
Antigen Presentation/genetics , Biolistics , Dendritic Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Transfection/immunology , Vaccines, DNA/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/biosynthesis , Antigens, CD/genetics , B7-2 Antigen , Cell Count , Dendritic Cells/metabolism , Female , Histocompatibility Antigens Class I/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin/cytology , Skin/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , Vaccines, DNA/administration & dosage , beta-Galactosidase/administration & dosage , beta-Galactosidase/genetics
8.
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
9.
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
10.
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
12.
J Immunol ; 156(9): 3357-65, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8617961

ABSTRACT

A number of cytokines and costimulatory molecules involved in immune activation have recently been identified including IL-12, a heterodimeric cytokine that supports the development of cell-mediated immunity, and B7-1, a costimulatory molecule involved in the activation of T lymphocytes. We explored the use of these immunomodulants as molecularly defined adjuvants in the function of recombinant anticancer vaccines using a murine model adenocarcinoma, CT26, transduced with a model Ag, beta-galactosidase (beta-gal). Although IL-12 given alone to mice bearing tumors established for 3 days did not have consistent antitumor activity, a profound therapeutic effect was observed when IL-12 administration was combined with a recombinant vaccinia virus (rVV) encoding beta-gal called VJS6. On the basis of the reported synergistic effects of IL-12 and the costimulatory molecule B7-1 (CD80) in vitro, we immunized mice with a double recombinant vaccinia encoding both the model tumor Ag and the costimulatory molecule B7-1, designated B7-1 beta-gal rVV. The adjuvant administration of IL-12 after immunization with this virus significantly enhanced survival in tumor-bearing animals. T cell subset depletions demonstrated that the in vivo activity of IL-12 was largely independent of CD4+ T lymphocytes, whereas the in vivo activity of a B7-1 rVV required both CD4+ and CD8+ T cells to elicit maximal therapeutic effect. To our knowledge, this is the first description of B7-1 and IL-12 cooperation in vivo and represents a novel strategy to enhance the efficacy of recombinant anticancer vaccines.


Subject(s)
Adjuvants, Immunologic/therapeutic use , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , Colonic Neoplasms/prevention & control , Interleukin-12/therapeutic use , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Adjuvants, Immunologic/antagonists & inhibitors , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/mortality , Female , Humans , Immunotherapy, Adoptive , Interleukin-12/antagonists & inhibitors , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Spleen/transplantation , Tumor Cells, Cultured , Vaccines, Synthetic/therapeutic use
13.
J Immunol ; 156(1): 238-45, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8598468

ABSTRACT

DNA immunization can result in the induction of Ag-specific cellular and humoral immune responses and in protective immunity in several Ag systems. To evaluate the utility of DNA-based immunization as a potential cancer treatment strategy, we employed an experimental murine tumor, CT26, expressing the model tumor-associated Ag, beta-galactosidase (beta-gal), designated CT26.CL25. A plasmid expressing beta-gal (pCMV/beta-gal) administered by particle-mediated gene delivery to the epidermis using a hand-held, helium-driven "gene gun" induced beta-gal-specific Ab and lytic responses. Immunization with this construct prevented the growth of pulmonary metastatic tumor, and the adoptive transfer of splenocytes generated by pCMV/beta-gal in vivo immunization and cultured in vitro with the beta-gal876-884 immunodominant peptide reduced the number of established pulmonary nodules. DNA immunization alone had little or no impact on the growth of established lung metastases. To enhance the function of DNA immunization for active immunotherapy, a panel of cytokines was added as adjuvants following DNA administration. Significant reduction in the number of established metastases was observed when human rIL-2, mouse rIL-6, human rIL-7, or mouse rIL-12 were given after DNA inoculation; mouse rIL-12 as an adjuvant had the most profound effect. These findings suggest that the cytokines involved in the activation and expansion of lymphocyte populations may improve the therapeutic effects of DNA immunization. Given the ease with which plasmid DNA can be prepared to high purity for safe use in humans with infectious diseases and cancers, DNA immunization administered together with cytokine adjuvant may be an attractive alternative to recombinant viral vaccines.


Subject(s)
Carcinoma/therapy , Colonic Neoplasms/therapy , Cytokines/therapeutic use , DNA, Neoplasm/therapeutic use , Genetic Therapy/methods , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Amino Acid Sequence , Animals , Carcinoma/genetics , Carcinoma/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , DNA, Neoplasm/administration & dosage , DNA, Neoplasm/immunology , Immunotherapy, Adoptive , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasm Transplantation , Tumor Cells, Cultured
14.
Semin Cancer Biol ; 6(6): 337-47, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8938272

ABSTRACT

The identification of tumor-associated antigens (TAA) recognized by T lymphocytes makes the development of antigen-specific synthetic and recombinant vaccines possible. The expression of TAA within a recombinant vector increases control over the kinetics and quantity, the molecular form, and the subcellular location of the immunogen delivered. The next generation of antitumor vaccines employs cytokines and costimulatory molecules expressed in concert with TAA that are capable of augmenting the activation and proliferation of antitumor immune responses. The ultimate goal of these new strategies, the treatment of established cancer, is now being realized in animal models.


Subject(s)
Antigens, Neoplasm/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology , Animals , Humans , Immunotherapy, Active , Neoplasms/therapy , T-Lymphocytes/immunology
15.
J Immunol ; 154(9): 4414-22, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7722298

ABSTRACT

CD8+ T lymphocytes (TCD8+) play an important role in cellular immune responses. TCD8+ recognize MHC class I molecules complexed to peptides of 8 to 10 residues derived largely from cytosolic proteins. Proteins are generally thought to be fragmented in the cytoplasm and delivered to nascent class I molecules in the endoplasmic reticulum (ER) by a peptide transporter encoded by the MHC. To explore the extent to which TCD8+ induction in vivo is limited by proteolysis or peptide transport into the ER, mice were immunized with recombinant vaccinia viruses containing mini-genes encoding antigenic peptides (bypassing the need for proteolysis), or these peptides with a NH2-terminal ER insertion sequence (bypassing the requirements for both proteolysis and transport). Additionally, mice were immunized with recombinant vaccinia viruses encoding rapidly degraded fragments of proteins. We report that limitations in induction of TCD8+ responses vary among Ags: for some, full length proteins are as immunogenic as other forms tested; for others, maximal responses are induced by peptides or by peptides targeted to the ER. Most importantly, in every circumstance examined, targeting peptides to the ER never diminished, and in some cases greatly enhanced, the TCD8+ immune response and provide an important alternative strategy in the design of live viral or naked DNA vaccines for the treatment of cancer and infectious diseases.


Subject(s)
Antigen Presentation/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Amino Acid Sequence , Animals , Cytotoxicity Tests, Immunologic , Female , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Orthomyxoviridae/immunology , Protein Sorting Signals/immunology
16.
J Immunol ; 154(9): 4651-7, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7722317

ABSTRACT

P815A is a naturally occurring tumor rejection Ag of the methylcholanthrene-induced murine mastocytoma P815. The gene encoding the Ag P815A, designated P1A, is identical to that encoded in the normal genome of the DBA/2 mouse. A recombinant vaccinia virus (rVV) was constructed that expressed a synthetic oligonucleotide encoding the minimal determinant peptide of the tumor-associated Ag. Although the rVV recombinant expressing this mini-gene was recognized efficiently in vitro, it was an ineffective immunogen in vivo. The addition of an endoplasmic reticulum insertion signal sequence to the NH2 terminus of the minimal determinant resulted in a rVV that elicited CD8+ T cells that could lyse P815 mastocytoma cells in vitro and that were therapeutic in vivo. Recombinant viruses expressing synthetic oligonucleotide sequences preceded by the insertion signal sequences allow the expression of Ag directly into the endoplasmic reticulum, where binding to MHC class I molecules is most efficient. Vaccines based on synthetic oligonucleotides could be constructed with ease and rapidity but, most importantly, such constructs avoid the dangers associated with the expression of full length genes encoding TAA that are potentially oncogenic.


Subject(s)
Mast-Cell Sarcoma/therapy , Oligonucleotides/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Base Sequence , Cytotoxicity Tests, Immunologic , Female , Mast-Cell Sarcoma/immunology , Mice , Mice, Inbred DBA , Molecular Sequence Data , Neoplasm Transplantation/immunology , Oligonucleotides/therapeutic use , Vaccines, Synthetic/therapeutic use , Vaccinia virus/genetics
17.
Cancer Res ; 55(8): 1741-7, 1995 Apr 15.
Article in English | MEDLINE | ID: mdl-7536130

ABSTRACT

Anticancer vaccine strategies can now target intracellular antigens that are involved in the process of malignant transformation, such as oncogene products or mutated tumor suppressor genes. Fragments of these antigens, generally 8-10 amino acids in length and complexed with MHC class I molecules, can be recognized by CD8+ T lymphocytes (TCD8+). To explore the possibility of using a genetically encoded, minimally sized fragment of an intracellular antigen as an immunogen, we constructed a recombinant vaccinia virus encoding an 8-residue peptide derived from chicken ovalbumin that is known to associate with the mouse H-2Kb molecule. Compared to standard methods of immunization, recombinant molecule. Compared to standard methods of immunization, recombinant vaccinia virus expressing the minimal determinant as well as full length ovalbumin were the only approaches that elicited specific primary lytic responses in C57BL/6 mice against E.G7OVA, a transfectant of the murine thymoma EL4 containing the ovalbumin gene. Stimulating these effectors in vitro with OVA257-264 peptide induced H-2Kb-restricted TCD8+ that not only lysed but also specifically secreted IFN-gamma in response to an antigen. Furthermore, when transferred adoptively, these anti-OVA257-264 TCD8+ cells significantly reduced the growth of established ovalbumin-transfected tumors in a pulmonary metastasis model system. Synthetic transfected tumors in a pulmonary metastasis model system. Synthetic oligonucleotides encoding minimal antigenic determinants within expression constructs may be a useful approach for treatment of neoplastic disease, thus avoiding the potential hazards of immunizing with full-length cDNAs that are potentially oncogenic.


Subject(s)
Cytotoxicity, Immunologic , T-Lymphocytes, Cytotoxic/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Vaccines, Synthetic/therapeutic use , Animals , Base Sequence , Chickens , Epitopes/biosynthesis , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligodeoxyribonucleotides , Ovalbumin/biosynthesis , Ovalbumin/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Thymoma/therapy , Thymus Neoplasms/therapy , Tumor Cells, Cultured , Vaccinia virus
18.
Folia Biol (Praha) ; 40(1-2): 74-88, 1994.
Article in English | MEDLINE | ID: mdl-7958066

ABSTRACT

Activated CD8+ T cells (TCD8+) can directly recognize malignant cells because processed fragments of tumour associated antigens (TAA), 8-10 amino acids in length and complexed with MHC class I molecules, are displayed on tumour cell surfaces. Tumour cells have been genetically modified in a variety of ways in efforts to enhance the immune recognition of TAA. An alternative strategy is the expression of TAA in recombinant or synthetic form. This has been made possible by the recent cloning of TAA recognized by TCD8+. In this communication we review recent work in our own laboratory on the expression of TAA as synthetic peptide, by "naked" plasmid DNA injected intramuscularly or transdermally, and by recombinant viruses including vaccinia (rVV), fowlpox (rFV) and adenovirus (rAd). The expression of TAA in recombinant and synthetic forms allows increased control over the quantity, location, and kinetics of TAA presentation and can result in powerful, specific, anti-tumour immune responses.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Viral , Immunotherapy , Interferon-gamma/genetics , Interferon-gamma/therapeutic use , Mice , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/genetics , Oligopeptides/immunology , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...