Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Int J Cancer ; 85(4): 508-17, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10699923

ABSTRACT

B7-1 is a co-stimulatory molecule that provides a second signal for T-cell activation. Several studies have demonstrated that vaccination with a vector containing genes encoding B7-1 and an antigen appears to be efficacious at promoting immune responsiveness to the antigen. To evaluate the safety of such a protocol and determine the effect of the B7-1 vector on immune responsiveness, female C57BL/6 mice were administered Wyeth wild-type vaccinia virus (V-WT) or V-WT containing the gene for B7-1 (rV-B7-1) as a single s. c. injection or 3 monthly s.c. injections. Immunologic parameters were evaluated in half of the mice and general toxicity in the other half. Immunologic end points included determination of splenic lymphocyte phenotypes, mitogen-induced T- and B-cell proliferation, T-cell proliferation in response to alloantigens, cell-mediated cytotoxicity (CMC), natural killer cell activity and serum anti-nuclear antibody (ANA) titers. No significant signs of general toxicity were noted. The primary immunologic effect was an increase in the ability of spleen cells to lyse allogeneic targets and to proliferate in response to allogeneic stimulation. Numbers of splenic CD8(+) cells were also increased. These effects were more pronounced after 3 vaccinations than after a single vaccination. Minimal differences in ANA were observed between mice immunized with V-WT and rV-B7-1. In addition, no serum antibodies against B7-1 were detected in any mice. The data suggest that vaccination with rV-B7-1 augments CMC with minimal toxicity.


Subject(s)
B7-1 Antigen/immunology , Cytotoxicity, Immunologic , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/toxicity , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Antibodies, Antinuclear/blood , B7-1 Antigen/genetics , Blood Cell Count , Blood Urea Nitrogen , Female , Immunity, Cellular , Mice , Mice, Inbred C57BL , Vaccinia virus
2.
Semin Oncol ; 26(6): 672-82, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606260

ABSTRACT

A new era involving the evaluation of recombinant vaccines for colon cancer has begun with the concurrent emergence of insights and technologies in the fields of molecular biology and immunology. These advances include (I) the identification and cloning of an array of genes associated with the neoplastic process, such as oncogenes, suppressor genes, genes encoding oncofetal antigens, and tissue lineage determinants; (2) the development of a variety of viral and bacterial vectors to deliver and present gene products; (3) the identification of numerous T-cell costimulatory molecules and the knowledge of their mode of action; (4) the cloning and analysis of the modes of action of an array of cytokines and other immunomodulatory molecules; and (5) a more sophisticated knowledge of the mode(s) of antigen presentation and T-cell activation.


Subject(s)
Cancer Vaccines , Colorectal Neoplasms/prevention & control , Adjuvants, Immunologic , Animals , Antigen Presentation , Antigens, Neoplasm , Carcinoembryonic Antigen , Clinical Trials as Topic , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Cytokines , Drug Evaluation, Preclinical , Epitopes , Gene Expression Regulation, Neoplastic , Genes, ras , Genetic Vectors , Humans , Lymphocyte Activation , Point Mutation , T-Lymphocytes
3.
Hum Gene Ther ; 10(7): 1095-103, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10340542

ABSTRACT

CD70, a ligand of the T cell costimulatory receptor CD27, is expressed mainly on activated B cells and has been shown to increase cytotoxic activity and proliferation of preferentially unprimed T cells. Reported herein is the construction of a recombinant vaccinia virus encoding CD70 (designated rV-CD70) and a demonstration of its biological effect on naive T cells in vitro and in vivo. In a whole tumor cell vaccine model, the growth of CD70-negative murine colon adenocarcinoma (MC38) tumor cells infected with rV-CD70 (multiplicity of infection [MOI] of 0.1) and transplanted into syngeneic C57BL/6 mice was inhibited completely while control tumors infected with wild-type vaccinia grew rapidly and killed mice within 3-5 weeks. Tumor-free mice previously immunized with rV-CD70-infected tumors were partially protected against rechallenge with wild-type tumors, demonstrating the induction of systemic anti-tumor immunity. In addition, immunization of C57BL/6 mice with rV-CD70 admixed with vaccinia virus encoding carcinoembryonic antigen (rV-CEA) was superior to treatment with rV-CEA alone in inducing CEA-specific lymphoproliferative T cell responses and reducing growth of murine colon carcinomas transduced with CEA. These studies demonstrate for the first time the potential utility of a recombinant vaccinia virus expressing CD70 to enhance T cell responses and mediate anti-tumor immunity.


Subject(s)
Adenocarcinoma/immunology , Antigens, CD , Cancer Vaccines/immunology , Colonic Neoplasms/immunology , Membrane Proteins/metabolism , Vaccinia virus/immunology , Animals , Antigen Presentation , CD27 Ligand , Cancer Vaccines/administration & dosage , Fluorescent Antibody Technique , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Transfection , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/transplantation , Tumor Cells, Cultured/virology , Vaccinia virus/genetics , Vaccinia virus/metabolism
4.
Hum Gene Ther ; 10(4): 623-31, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10094205

ABSTRACT

T cell activation requires binding of the T cell receptor to the major histocompatibility molecule-peptide complex in the presence of adhesion and/or costimulatory molecules such as B7-1 (CD80), B7-2 (CD86), ICAM-1 (CD54), and LFA-3 [corrected]. The major ligand of CD2 is CD48, the murine analog of human leukocyte function-associated antigen 3 (LFA-3). To determine the effect of LFA-3 expression on the immunogenicity of tumor cells, we constructed a recombinant vaccinia virus containing the murine LFA-3 gene (designated rV-LFA-3). rV-LFA-3 was shown to be functional in vitro in terms of expression of LFA-3, T cell proliferation, adhesion, and cytotoxicity. Subcutaneous inoculation of rV-LFA-3-infected murine colon adenocarcinoma tumor cells (MC38) into immunocompetent syngeneic C57BL/6 mice resulted in complete lack of tumor growth. Inoculation of MC38 cells infected with equal doses of control wild-type vaccinia virus resulted in tumor growth in all animals. In addition, partial immunological protection was demonstrated against subsequent challenge with uninfected parental tumor cells up to 56 days after vaccination with rV-LFA-3-infected cells. Anti-tumor memory was also demonstrated by using gamma-irradiated MC38 cells and cells from another carcinoma model (CT26). These studies demonstrate that expression of LFA-3 via a poxvirus vector can be used to induce anti-tumor immunity.


Subject(s)
Adenocarcinoma/immunology , CD58 Antigens/genetics , Colonic Neoplasms/immunology , Recombination, Genetic , Vaccinia virus/genetics , Adenocarcinoma/pathology , Animals , Base Sequence , Cell Division/immunology , Colonic Neoplasms/pathology , DNA Primers , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology
5.
Vaccine ; 17(7-8): 893-903, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10067696

ABSTRACT

Several recombinant vaccinia viruses are currently being evaluated to induce antigen-specific immunity to a variety of infectious disease agents and tumor associated antigens. T-cell costimulation is extremely important in enhancing T-cell responses, and recombinant vaccines have now been shown to be effective vectors to express a range of these molecules. Both combination vaccines (an admixture of a recombinant vaccinia virus expressing a specific target antigen and a recombinant vaccinia virus expressing a costimulatory molecule) and dual-gene vaccines expressing both transgenes on the same vector have been shown capable of effectively enhancing antigen-specific responses via T-cell costimulation. In this report, we compare for the first time the use of both types of approaches to enhance antigen-specific T-cell responses, and we demonstrate the importance of route of vaccine administration and vaccine dose in attaining optimal T-cell responses. These studies should have direct bearing on the design of vaccine clinical trials for infectious agents and/or tumor associated antigens, in which T-cell costimulatory molecules will be employed to enhance antigen-specific T-cell responses via the use of either combination or dual-gene vaccinia vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/genetics , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Epitopes, T-Lymphocyte/immunology , Female , Immunity, Cellular/immunology , Mice , Mice, Inbred C57BL , Transgenes/immunology , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, DNA/genetics , Vaccinia virus/genetics , Viral Vaccines/genetics , Viral Vaccines/pharmacology
6.
Cell Immunol ; 187(2): 103-16, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9732698

ABSTRACT

Point mutations in the ras proto-oncogenes, notably at codon 12, are found in high frequency of human malignancies and, thus, may be appropriate targets for the induction of tumor-specific T cell responses in cancer immunotherapy. In this study, we examined the mutant ras protein sequence reflecting the substitution of Gly to Val at position 12 as a putative point-mutated determinant for potential induction of an HLA-A2-reactive, CD8+ cytotoxic T lymphocyte (CTL) response. We identified the ras 4-12(Val12) sequence as a minimal 9-mer peptide, which displayed specific binding to HLA-A2 by T2 bioassays. Peptide binding to HLA-A2 on T2 cells was weak and required coincubation with exogenous beta(2)-microglobulin to facilitate and enhance complex formation. In contrast, the wild-type ras 4-12(Gly12) peptide failed to bind to HLA-A2 even in the presence of beta(2)-microglobulin, consistent with the hypothesis that the point mutation creates a C-terminus anchor residue. A CD8+ CTL line against the ras 4-12(Val12) peptide was derived in vitro from a normal HLA-A2+ donor using a model culture system consisting of T2 cells as antigen presenting cells pulsed with exogenous mutant ras peptide and beta(2)-microglobulin plus cytokines (interleukin-2 and 12). Functional characterization of CD8+ CTL line revealed (1) peptide-specific and HLA-A2-restricted cytotoxicity against a panel of peptide-pulsed targets; (2) no specific lysis using the normal ras peptide sequence; (3) half-maximal lysis with exogenous peptide of approximately 0.3 microM; (4) lysis of HLA-A2+ B cell lines infected with a recombinant vaccinia virus construct encoding the point-mutated human K-ras gene; and (5) specific lysis of the HLA-A2+ SW480 colon carcinoma cell line expressing the naturally occurring K-ras Val12 mutation. Maximal lysis of SW480 cells occurred following interferon (IFN)-gamma pretreatment, which correlated with enhanced HLA-A2 and ICAM-1 (CD54) expression. Specificity of lysis was revealed by the absence of lysis against a HLA-A2+ melanoma cell line (+/- IFN-gamma), which lacked the mutant Val12 mutation, and the inability of an irrelevant CD8+ CTL line to lyse SW480 (+/- IFN-gamma) unless the appropriate exogenous peptide was added. These findings demonstrated that tumor cells may endogenously process and express mutant ras epitopes, such as the 4-12(Val12) sequence, albeit in limiting amounts that may be potentiated by IFN-gamma treatment. These data support the biological relevance of this sequence and, thus, may have important implications for the generation of ras oncogene-specific CTL responses in clinical situations.


Subject(s)
Alleles , Epitopes, T-Lymphocyte , HLA-A2 Antigen/genetics , Point Mutation , T-Lymphocytes, Cytotoxic/immunology , ras Proteins/immunology , Codon , Genes, ras , Humans , Peptide Fragments/immunology
7.
Expert Opin Investig Drugs ; 7(9): 1439-52, 1998 Sep.
Article in English | MEDLINE | ID: mdl-15992042

ABSTRACT

A new era involving the evaluation of recombinant cancer vaccines has begun with the concurrent emergence of insights and technologies in the fields of molecular biology and immunology. These advances include: The identification and cloning of an array of genes associated with the neoplastic process, such as oncogenes, suppressor genes, genes encoding oncofoetal antigens and tissue-lineage determinants. The development of a variety of viral and bacterial vectors to deliver and present gene products. The identification of numerous T-cell costimulatory molecules and an understanding of their mode of action. The cloning and analysis of the modes of action of an array of cytokines and other immunomodulatory molecules. More sophisticated knowledge of the mode(s) of antigen presentation and T-cell activation. One current challenge in cancer therapy is the delineation of strategies toward the rational design and implementation of recombinant vaccines that will be of therapeutic benefit to cancer patients and/or members of groups at high risk for specific neoplasias. Numerous concepts are emerging in this regard. The study of immunologic intervention using laboratory animal models demonstrates that no one approach will prevail for all cancer types or, perhaps, for the various stages of the neoplastic process of a given tumour type. The immunological role(s) of CD8+, CD4+, natural killer and other cell types, as well as the roles of antibodies, must all be taken into consideration. This article reviews some of the strategies currently undergoing evaluation toward the development of recombinant vaccines for several carcinoma types.

8.
Hum Gene Ther ; 8(7): 851-60, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9143911

ABSTRACT

The intercellular adhesion molecule-1 (ICAM-1) has been associated with cellular migration into inflammatory sites and with facilitating interactions between lymphocytes and tumor targets in the pathway of cell-mediated cytotoxicity. More recently, ICAM-1 has become increasingly implicated in the costimulation of T cell functions, such as antigen-dependent T cell proliferation. Previous murine studies have shown that the introduction of the ICAM-1 gene into tumor cells using retroviral vectors led to enhanced antitumor responses. In this study, we report the construction, characterization, and immunological consequences of a recombinant vaccinia virus expressing murine ICAM-1. Vaccinia virus represents an attractive vector for the delivery of molecules such as ICAM-1 due to its wide host range, rapid infection, and functional expression of inserted gene products. The infection of tumor cells with this recombinant virus resulted in the expression of functional ICAM-1. Infected tumors provide accessory or secondary signals to lymphoblasts in vitro, resulting in enhanced cytokine production or alloreactive cytotoxic T lymphocyte (CTL) activity. In vivo, we demonstrated that weakly immunogenic syngeneic tumors, infected with and expressing rV-ICAM-1, were rejected by immunocompetent hosts. Furthermore, immunization with rV-ICAM-1-infected tumors resulted in the rejection of subsequent tumor challenge, providing evidence for recall response and immunological memory. These studies demonstrated the utility of a recombinant vaccinia virus to deliver and efficiently express ICAM-1 molecules on tumor cells for potential gene therapy and recombinant approaches to cancer immunotherapy.


Subject(s)
Genetic Vectors , Intercellular Adhesion Molecule-1/genetics , Vaccines, Synthetic/genetics , Vaccinia virus/genetics , Adenocarcinoma/prevention & control , Animals , Base Sequence , Colonic Neoplasms/prevention & control , DNA , Female , Gene Expression , Intercellular Adhesion Molecule-1/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombination, Genetic , Tumor Cells, Cultured , Vaccines, Synthetic/immunology
9.
Vaccine ; 15(6-7): 759-68, 1997.
Article in English | MEDLINE | ID: mdl-9178479

ABSTRACT

Recombinant vaccinia viruses containing tumor associated genes represent an attractive vector to induce immune responses to weak immunogens in cancer immunotherapy protocols. The property of intense immunogenicity of vaccinia proteins, however, also serves to limit the number of inoculations of recombinant vaccinia viruses. Host immune responses to the first immunization have been shown to limit the replication of subsequent vaccinations and thus reduce effectiveness of boost inoculations. The use of recombinant avian pox viruses (avipox) such as the canarypox (ALVAC) or fowlpox are potential candidates for immunization protocols in that they can infect mammalian cells and express the inserted transgene, but do not replicate in mammalian cells. We report here the construction and characterization of a canarypox (ALVAC) recombinant expressing the human carcinoembryonic antigen (CEA) gene (designated ALVAC-CEA). Antibody, lymphoproliferative and cytolytic T-cell responses as well as tumor inhibition were shown to be elicited by the ALVAC-CEA recombinant in a murine model. The utilization of a diversified immunization scheme using a recombinant vaccinia virus followed by recombinant avian pox virus was shown to be far superior than the use of either one alone in eliciting CEA-specific T-cell responses. Experiments were conducted to determine if the use of a diversified immunization scheme using a recombinant vaccinia virus (rV-CEA) and ALVAC-CEA would be superior to the use of either one alone in eliciting CEA-specific T-cell responses. When mice were immunized with rV-CEA and then ALVAC-CEA. CEA-specific T-cell responses were at least four times greater, and for superior to those achieved with three immunizations of ALVAC-CEA. Multiple boosts of ALVAC-CEA following rV-CEA immunization further potentiated anti-tumor effects and CEA specific T-cell responses. These studies demonstrate the proof of concept of the advantage of diversified immunization protocols employing both recombinant vaccinia and recombinant avipox vectors.


Subject(s)
Adenocarcinoma/prevention & control , Avipoxvirus , Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Vaccinia virus , Animals , Antibodies, Neoplasm/analysis , Avipoxvirus/physiology , Carcinoembryonic Antigen/genetics , Cell Line , Chlorocebus aethiops , Cytotoxicity Tests, Immunologic , Female , Genetic Vectors , Humans , Immunization , Immunization, Secondary , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Virus Replication
10.
J Immunother ; 20(1): 38-47, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9101412

ABSTRACT

Tumor-associated antigens have considerable promise not only as diagnostic or prognostic markers but also as targets for active or passive immunotherapy. DF3/MUC1 is a tumor-associated antigen that is overexpressed with an abnormal glycosylation pattern in breast, ovarian, lung, and pancreatic cancers. The major extracellular portion of MUC1 is composed of tandem repeat units of 20 amino acids. Recombinant vaccinia viruses encoding mucin molecules have been constructed by several groups. However, these recombinants have met with limited success in protecting animals from MUC1-expressing tumors because of the vaccinia genome being subject to high-frequency homologous recombination, therefore being unstable in expression of the tandem repeats. In light of these studies, two concurrent strategies were used to improve immune responses to MUC1: a recombinant vaccinia virus was constructed containing a modified "mini" MUC1 gene containing only 10 tandem repeat sequences to minimize vaccinia-mediated rearrangement (designated rV-MUC1); and an admixture was used containing rV-MUC1 and a recombinant vaccinia virus containing the gene for the murine T-cell costimulatory molecule B7-1 (rV-B7). The rV-MUC1 gene product maintained a consistent molecular weight throughout several passages, indicating stability of the inserted gene. Mice inoculated with rV-MUC1 demonstrated MUC1-specific cytolytic responses that were further enhanced by admixture with rV-B7. In a MUC1-expressing pulmonary metastases prevention model, mice inoculated two times with rV-MUC1 were protected from the establishment of metastases. No additive effect on antitumor immunity (> 90% with rV-MUC1 alone) was observed in mice primed with an admixture of rV-MUC1 and rV-B7 and boosted with rV-MUC1. When rV-MUC1 was used to treat established MUC1 positive metastases, however, three administrations of rV-MUC1 were not sufficient to confer antitumor effects. In contrast, when tumor-bearing mice were primed with an admixture of rV-MUC1 and rV-B7, followed by two boosts with rV-MUC1, there was a significant reduction in pulmonary metastases (p = < 0.0001), which correlated to 100% survival. Coexpression of the B7 molecule, although not necessary for the induction of an immune response of sufficient magnitude to prevent MUC1 tumors, was thus essential in a treatment setting.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/therapeutic use , Gene Expression Regulation, Viral/physiology , Genes, Viral , Vaccines, Synthetic/therapeutic use , Vaccinia virus/genetics , Animals , B7-1 Antigen/genetics , Cancer Vaccines/genetics , Cytotoxicity Tests, Immunologic , Mice , Neoplasm Metastasis/prevention & control , Recombination, Genetic , Repetitive Sequences, Nucleic Acid , T-Lymphocytes/immunology , Transduction, Genetic , Tumor Cells, Cultured , Vaccines, Synthetic/genetics
11.
J Immunother ; 20(1): 48-59, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9101413

ABSTRACT

We have described previously the construction, generation, and in vivo biologic consequences of a recombinant vaccinia virus containing the human CEA gene (rV-CEA) in an experimental murine colon carcinoma model. Immunization of C57BL/6 mice with rV-CEA led to antigen-specific inhibition of tumor growth in both prophylactic and therapeutic settings. Although such antitumor effects were correlated with the induction of CEA-specific T-cell responses, their exact contribution in the tumor rejection mechanism remained unclear. In this study, we examined the mechanism of action of rV-CEA, with emphasis on definition of the immune cells important for such antitumor effects. To that end, a cellular adoptive transfer model was established in vivo, which allowed specific functional analysis of donor-derived immune cells in naive, sublethally irradiated, tumor-bearing recipients. Splenocytes from rV-CEA-immunized donors expressed strong antitumor activity in such tumor-bearing recipients, whereas nonimmune donor cells did not. Depletion of immune T cells before cellular transfer abolished the antitumor response. Moreover, depletion of CD8+ T cells before transfer resulted in the loss of antitumor activity, despite the presence of CD4+ T cells. In contrast, antitumor activity was demonstrable with CD8-containing, CD4-depleted effectors, although it was not as effective as with both T-cell subpopulations combined. Finally, in beta 2-microglobulin/CD8+ T-cell-deficient mice, rV-CEA immunization exerted only partial antitumor protection, compared with the immune-competent controls. Overall, we demonstrated that (a) antitumor activity induced by rV-CEA was essentially mediated by CD8+ effectors; and (b) the combination of both CD8+ and CD4+ lymphocytes led to maximal antitumor therapeutic effects, suggesting an important helper or immunoregulatory contribution of the CD4+ subset. Thus, adoptive cellular transfer strategies may have implications for both the study of recombinant anticancer vaccines and the development of potential clinical applications for cancer immunotherapy.


Subject(s)
Adenocarcinoma/therapy , Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/genetics , Colonic Neoplasms/therapy , Immunotherapy, Adoptive , Vaccines, Synthetic/therapeutic use , Animals , Antibody Specificity , CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombination, Genetic , Spleen/cytology , Spleen/immunology
12.
Cell Immunol ; 182(2): 137-51, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9514698

ABSTRACT

Previous studies have identified and characterized both murine in vivo and human in vitro T cell responses reflecting specific mutations in the ras proto-oncogenes at codon 12, 13, or 61. In an attempt to determine whether peptide epitopes reflecting point mutations in the ras oncogenes are immunogenic in humans for the production of CD4+ and/or CD8+ T cell responses, a phase I clinical trial was initiated in metastatic carcinoma patients whose primary tumors harbor mutations in the K-ras proto-oncogenes at codon 12. The peptides used here as immunogens, which were administered in Detox adjuvant, spanned the ras sequence 5-17 and reflected the amino acid substitution of glycine (Gly) at position 12 to aspartic acid (Asp), cysteine (Cys), or valine (Val). Three of eight evaluable patients have demonstrated peptide-specific cell-mediated immunity, as determined by the production of T cell lines resulting from the vaccination. First, an antigen (Ag)-specific, major histocompatibility complex (MHC) class II (DP)-restricted CD4+ T cell line was established in vitro from postvaccination lymphocytes of a non-small cell lung carcinoma patient whose primary tumor contained a Cys12 mutation when cultured on the immunizing peptide. Moreover, CD4+ proliferation was inducible against the corresponding mutant K-ras protein, suggesting productive T cell receptor recognition of exogenously processed Ag. Second, an Ag-specific, MHC class I (HLA-A2)-restricted CD8+ cytotoxic T lymphocyte (CTL) line was established in vitro from postvaccination lymphocytes of a colon carcinoma patient whose primary tumor contained an Asp12 mutation. To that end, a 10-mer peptide, nested within the 13-mer immunizing peptide, was identified [i.e., ras5-14(Asp12)], which was shown to bind to HLA-A2 and display specific functional capacity for expansion of the in vivo primed CD8+ CTL precursors. Third, both Ag-specific, MHC class II (DQ)-restricted CD4+ and MHC class I-restricted (HLA-A2) CD8+ T cell lines were generated from a single patient with duodenal carcinoma whose primary tumor contained a Val12 mutation when cultured on the immunizing 13-mer peptide or a nested 10-mer peptide [i.e., ras5-14(Val12)], respectively. Evidence is thus provided that vaccination with mutant ras oncogene peptides in adjuvant may induce specific anti-ras cellular immune responses, with no detectable cross-reactivity toward normal proto-ras sequences. Moreover, we have identified for the first time human HLA-A2-restricted, CD8+ CTL epitopes reflecting specific point mutations in the K-ras oncogenes at codon 12 which, in concert with the activation of the CD4+ T cell response, may have important implications for both active and passive immunotherapies in selected cancer patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, ras , Point Mutation , Adult , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Codon/genetics , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , HLA-DQ Antigens/metabolism , Humans , Immunization , In Vitro Techniques , Lymphocyte Activation , Male , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , ras Proteins/genetics , ras Proteins/immunology , ras Proteins/metabolism
13.
Cancer Res ; 56(10): 2361-7, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8625312

ABSTRACT

Interleukin-2 (IL-2) has been an effective immune modulator in several active-specific immunotherapy experimental protocols using either viral or oncolysate-based vaccines. In this report, data indicate that IL-2 administration can appreciably augment the therapeutic effect of a single immunization of a recombinant vaccinia virus-carcinoembryonic antigen (rV-CEA) vaccine using a CEA-expressing syngeneic experimental murine model system. A single rV-CEA immunization of C57BL/6 mice bearing palpable CEA-positive colon adenocarcinoma tumors results in complete tumor regression in approximately 20% of the mice. The addition of a course of low-dose IL-2 results in complete tumor regression in 60-70% of the mice. Moreover, the combination of rV-CEA and IL-2 induces systemic immunity, which protects those tumor-free mice from subsequent rechallenge with the CEA-expressing tumor cells. No such tumor regression or protection was observed in those mice immunized with the wild-type vaccinia vaccine (V-Wyeth) alone or with IL-2 administration alone. Cellular immune assays revealed that the addition of IL-2 to rV-CEA immunization significantly increased the CEA-specific T-cell proliferative responses as well as the cytolytic T-cell responses when compared with rV-CEA immunization alone. The enhanced CEA-specific immune response, coupled with the improved experimental therapeutic outcome following IL-2 administration, suggests that treatment with that cytokine may effectively substitute for multiple rV-CEA immunizations in active-specific immunotherapy clinical protocols directed at CEA-expressing tumors.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antigens, Neoplasm/immunology , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/therapy , Immunotherapy, Active , Interleukin-2/therapeutic use , Recombinant Fusion Proteins/immunology , Smallpox Vaccine/administration & dosage , Vaccinia virus/immunology , Adjuvants, Immunologic/pharmacology , Animals , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Female , Humans , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Smallpox Vaccine/immunology , Smallpox Vaccine/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
14.
Int J Cancer ; 63(2): 231-7, 1995 Oct 09.
Article in English | MEDLINE | ID: mdl-7591210

ABSTRACT

Prostate-specific antigen (PSA) is a serine protease secreted by prostatic epithelial cells and is widely used as a marker for prostate cancer. The tissue specificity of PSA makes it a potential target for active specific immunotherapy, especially in prostate cancer patients who have undergone prostatectomy and in whom the only PSA-expressing tissue in the body resides in metastatic deposits. We report here the cloning, construction and immunological consequences of immunization of rhesus monkeys with a recombinant vaccinia virus expressing human PSA (designated rV-PSA). The prostate gland of the rhesus is structurally and functionally similar to the human prostate. While rodent and other mammalian species do not share homology with human PSA, there is 94% homology between the amino acid sequences of rhesus and human PSA. Immunization of rhesus monkeys with wild-type vaccinia virus or rV-PSA elicited the usual low-grade constitutional symptoms of vaccinia virus infection. There was no evidence of any adverse effects in any immunized monkeys. A short-lived PSA-specific IgM antibody response was noted in all rV-PSA immunized monkeys regardless of dose level. All monkeys receiving the 10(8)pfu dose of rV-PSA demonstrated PSA-specific T-cell responses that were maintained up to 270 days. No differences in anti-PSA immune responses or toxicity were observed in animals that received prostatectomy prior to immunization. Our results thus demonstrate the safety and immunogenicity of rV-PSA in a non-human primate and have implications for potential specific immunotherapy protocols using PSA as a target.


Subject(s)
Prostate-Specific Antigen/immunology , Animals , Antibodies, Neoplasm/immunology , Base Sequence , Cloning, Molecular , DNA Primers/chemistry , DNA, Complementary/genetics , Humans , Immunoglobulin M/immunology , Lymphocyte Activation , Macaca mulatta , Molecular Sequence Data , Prostate-Specific Antigen/genetics , Recombinant Proteins , Vaccines, Synthetic/immunology , Vaccinia virus
15.
Eur J Immunol ; 25(9): 2588-97, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7589131

ABSTRACT

Alterations in the ras p21 protein have been associated with both rodent and human neoplasia. Thus, mutated ras p21 proteins may bear unique antigenic epitopes for immune recognition, such as by T cells, which have been implicated in host antitumor activity. Synthetic peptides that mimic segments of mutated ras p21 have been reported to be immunogenic in mice in vivo, although detailed functional analyses remains undefined. Here, in a murine model, we explored and characterized distinct effector properties of host-derived T lymphocytes reactive to mutated ras peptides, which was consistent with the CD4+ T helper type 1 (Th1) subset. BALB/c mice (H-2d) were immunized with a purified peptide, 13 amino acids in length, containing the substitution of Gly (G12) to Val (V12) at position 12, which is commonly found in human carcinomas. An alpha beta T cell receptor-positive, CD3+, CD4+, CD8- T cell line was established, which expressed peptide-specific proliferation. Cytokine assays revealed the production of interleukin-2, interferon-gamma, tumor necrosis factor and granulocyte-macrophage colony-stimulating factor. Moreover, antigen-specific cytotoxicity was demonstrable against: (1) Iad-bearing A20 tumor cells incubated with exogenously bound V12 peptide; and (2) A20 tumor cells transduced with the K-ras p21 oncogene encoding the corresponding point mutation. CD4(+)-mediated cytotoxicity was major histocompatibility complex (MHC) class II-restricted, as revealed by the absence of lysis against MHC class II- P815 targets, inhibition of A20 lysis with anti-Iad monoclonal antibodies, and induction of lysis against L cell targets transfected with E alpha A beta d. Independent isolation of a second CD4+ V12 line revealed a very similar cytolytic and MHC class II-restricted profile. Overall, these data demonstrated that peptide immunization produced a CD4+ Th1 response that specifically recognized tumor cells expressing endogenous activated K-ras epitopes, which may have implications for the development of peptide-based active immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Oncogene Protein p21(ras)/immunology , Animals , Base Sequence , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Oncogene Protein p21(ras)/genetics , Point Mutation , Tumor Cells, Cultured
16.
Cancer Res ; 55(16): 3598-603, 1995 Aug 15.
Article in English | MEDLINE | ID: mdl-7543017

ABSTRACT

At least two signals are required for the activation of naive T cells by antigen-bearing target cells: an antigen-specific signal, delivered through the T-cell receptor, and a costimulatory signal delivered through the T-cell surface molecule CD28 by its natural ligand B7-1. The immunological benefit of coexpression of B7 with target antigen has been demonstrated with the use of several retroviral systems to transfect antigen-bearing cells. Although engineering recombinant constructs with genes for two or more antigens can mediate the dual expression of those antigens, disadvantages of this approach include the time for construction of each desirable combination and the inability to control differential expression levels of each gene product. An alternative approach would utilize separate constructs that could be admixed appropriately before administration. In this report we describe the functional consequences of the admixture of recombinant vaccinia murine B7-1 (rV-B7) to recombinant vaccinia expressing the human carcinoembryonic antigen gene (rV-CEA). Coinfection of cells resulted in high levels of cell surface expression of both the CEA and B7 molecules. Immunization of mice with various ratios (1:3, 1:1, 3:1) of rV-CEA and rV-B7 demonstrated that an admixture of rV-CEA and rV-B7 at a 3:1 ratio resulted in the generation of optimal CEA-specific T-cell responses. Next, we examined the efficacy of this admixture on antitumor activity. Typically, injection of murine carcinoma cells expressing CEA leads to the death of the host. One immunization of C57BL/6 mice with rV-CEA:rV-B7 (3:1) resulted in no tumor establishment. In contrast, administration of rV-CEA or rV-B7 alone had little or no antitumor effects. These studies demonstrate the advantages of the use of recombinant vaccinia viruses to deliver B7 molecules in combination with a tumor-associated antigen. The availability of the rV-B7 single construct and the ability to alter the B7 ratio could also have potential utility when coinfecting rV-B7 with recombinant vaccinia viruses containing genes for infectious agents or other tumor-associated antigen genes.


Subject(s)
B7-1 Antigen/immunology , Carcinoembryonic Antigen/immunology , Neoplasms, Experimental/prevention & control , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes/immunology , Animals , B7-1 Antigen/genetics , Carcinoembryonic Antigen/genetics , Cytotoxicity, Immunologic , Female , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Recombinant Proteins , Vaccines, Synthetic , Vaccinia virus
17.
Cancer Res ; 55(11): 2455-62, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7538903

ABSTRACT

Human prostate-specific antigen (PSA) has been shown as an aid in the early detection of prostate cancer (W. J. Catalona et al., J. Am. Med. Assoc., 270: 948-954, 1993) and was approved in 1994 by the Food and Drug Administration for early detection of prostate cancer. Immunotherapies directed against PSA have been suggested in patients with metastatic prostate cancer. One of the essential questions is to define which nonhuman species express PSA for experimental studies. Using Southern blot analyses, genes related to human PSA have been detected in several nonhuman primate species, including chimpanzee, orangutan, gorilla, macaque, and rhesus monkey, but not in other mammalian species, including rabbit, cow, pig, dog, rat, or mouse. Immunohistochemical staining with anti-human PSA antisera detected strong staining in both human and monkey prostatic epithelial cells with no reactivity to rat prostate cells. Because the PSA gene is not present in the murine genome, a matched set of murine cell lines has been developed that may be useful to study the biochemical functions of PSA and as an experimental target for PSA-directed immunotherapy. To establish such cell lines, a C57BL/6 murine colon adenocarcinoma cell line, MC-38, was transfected with a retroviral vector containing cDNA encoding the human PSA gene. Genetic analysis of a PSA-secreting clone, PSA/MC-38, demonstrated that the PSA gene had been stably integrated into the MC-38 genome. The PSA/MC-38 cell line was found to secrete PSA into tissue culture medium, producing a protein of approximately M(r) 30,000. In vivo, PSA/MC-38 grew as a s.c. tumor in male and female mice. PSA/MC-38 tumors grew more rapidly in athymic mice than in syngeneic C57BL/6 mice, and in both mouse strains, the PSA/MC-38 tumors grew more slowly than control vector-transduced tumors. PSA was detected in the serum and tumors of PSA/MC-38 tumor-bearing mice. It is proposed that PSA/MC-38 cells may be used as a murine tumor model to test potential therapeutic vaccines and other experimental therapies directed against PSA.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Adenocarcinoma/chemistry , Animals , Base Sequence , Cell Division/physiology , Colonic Neoplasms/chemistry , DNA, Neoplasm/genetics , Disease Models, Animal , Female , Gene Expression , Genome , Humans , Immunohistochemistry , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Sequence Data , Prostate/chemistry , Prostate/metabolism , Prostate/physiology , Prostate-Specific Antigen/analysis , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/chemistry , Rats , Rats, Wistar , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
18.
Cancer Res ; 54(21): 5552-5, 1994 Nov 01.
Article in English | MEDLINE | ID: mdl-7522961

ABSTRACT

Activation of T cells requires at least two signals: an antigen-specific signal delivered through the T-cell receptor and a costimulatory signal mediated through molecules designated B7-1 and B7-2. Previous studies have shown that introduction of B7-1 and B7-2 into tumors using retroviral vectors has led to enhanced antitumor effects. A limiting factor for potential clinical applications using this approach is the low efficiency of infection of retroviral vectors and consequent manipulations of infected cells. Vaccinia virus thus represents an alternative vector for B7 gene expression in tumor cells. In this report we describe the construction and characterization of recombinant vaccinia viruses containing the murine B7-1 and B7-2 genes (designated rV-B7-1 and rV-B7-2). Infection of BSC-1 cells with these constructs results in rapid and efficient cell surface expression of both B7-1 and B7-2 (> 97% of cells at 4 h). Infection of murine carcinoma cells with low multiplicity of infection of wild-type vaccinia virus leads to the death of the host following tumor transplantation. In contrast, inoculation of rV-B7-1- or rV-B7-2-infected tumor cells into immunocompetent animals resulted in no tumor growth. These studies demonstrate the utility of recombinant vaccinia viruses to deliver B7 molecules to tumor cells for potential gene therapy and recombinant approaches to cancer immunotherapy.


Subject(s)
Antigens, CD , B7-1 Antigen/immunology , Membrane Glycoproteins , Vaccinia virus/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , B7-1 Antigen/metabolism , B7-2 Antigen , Base Sequence , Cell Division , DNA, Complementary/genetics , DNA, Viral/genetics , Female , Genetic Vectors , Immunocompetence , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Transplantation , Recombination, Genetic , Tumor Cells, Cultured , Vaccinia virus/genetics , Vaccinia virus/metabolism
19.
Cancer Res ; 51(14): 3657-62, 1991 Jul 15.
Article in English | MEDLINE | ID: mdl-1712245

ABSTRACT

A cell line derived from the mouse colon adenocarcinoma, MC-38, has been transduced with a retroviral construct containing complementary DNA encoding the human carcinoembryonic antigen (CEA) gene. MC-38, which forms tumors in syngeneic C57BL/6 mice, has been extensively studied as a target for active immunotherapy. Individual transduced clones that express high levels of cell surface CEA were isolated, and two clones, termed MC-38-ceal and MC-38-cea2, were extensively characterized. The levels of CEA found on the surface of these clones were considerably higher than that found in a moderately differentiated human colon carcinoma cell line (WiDr) and were comparable to those found on the human colon carcinoma cell lines GEO and CBS (among the highest CEA-expressing cells reported). Further analysis demonstrated that the CEA expressed in the MC-38-cea1 clone had a similar molecular weight to native CEA (Mr 180,000), but the MC-38-cea2 cell line expressed a single Mr 70,000 glycosylated immunoreactive product. Seven anti-CEA monoclonal antibodies were found to react with both clones. The CEA gene present in the MC-38-cea2 clone was partially sequenced and was found to contain a deletion of two of the three repeated domains present in CEA. These results provide a basis for future studies to map immunodominant epitopes of CEA and to develop a syngeneic model system that may aid in the design of reagents and protocols to study active and passive immunotherapy directed against a carcinoma expressing human CEA.


Subject(s)
Adenocarcinoma/immunology , Antigens, Neoplasm , Carcinoembryonic Antigen/genetics , Cell Adhesion Molecules , Colonic Neoplasms/immunology , Gene Expression , Transduction, Genetic , Adenocarcinoma/genetics , Animals , Base Sequence , Carcinoembryonic Antigen/analysis , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/genetics , Epitopes/analysis , Membrane Glycoproteins/immunology , Mice , Molecular Sequence Data , Molecular Weight , Tumor Cells, Cultured
20.
Proc Natl Acad Sci U S A ; 80(8): 2318-22, 1983 Apr.
Article in English | MEDLINE | ID: mdl-6572978

ABSTRACT

A beta+-thalassemia globin gene was isolated from the genome of a Black individual by molecular cloning. DNA sequence analysis revealed only a single difference between this gene and the normal human beta-globin gene--adenine is substituted for thymine in the third position of codon 24. Codon 24 in both the normal gene (GGT) and the beta+-thalassemia gene (GGA) encodes glycine. The function of this beta+-thalassemia gene was compared to the function of the normal human beta-globin gene in monkey kidney cells by using plasmid expression vectors. The codon 24 substitution activates a 5' splice site that involves the guanine-thymine dinucleotide present in codon 25, 16 nucleotides upstream from the normal exon 1-intron I boundary. The splice, involving the abnormal 5' site in codon 25, is completed with the normal 3' splice site at the end of intron I. This splicing abnormality leads to a 75% decrease in the accumulation of normally processed beta-globin mRNA, thereby causing the beta+-thalassemia phenotype.


Subject(s)
Globins/genetics , RNA Splicing , RNA, Messenger/genetics , Thalassemia/genetics , Base Sequence , Cloning, Molecular , Codon , Gene Expression Regulation , Humans , RNA Processing, Post-Transcriptional
SELECTION OF CITATIONS
SEARCH DETAIL
...