Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cancer Immunol Immunother ; 62(2): 371-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22926061

ABSTRACT

The central role of CD4+ T lymphocytes in mediating DNA vaccine-induced tumor immunity against the viral oncoprotein simian virus 40 (SV40) large tumor antigen (Tag) has previously been described by our laboratory. In the present study, we extend our previous findings by examining the roles of IFN-γ and Th1-associated effector cells within the context of DNA immunization in a murine model of pulmonary metastasis. Immunization of BALB/c mice with plasmid DNA encoding SV40 Tag (pCMV-Tag) generated IFN-γ-secreting T lymphocytes that produced this cytokine upon in vitro stimulation with mKSA tumor cells. The role of IFN-γ as a mediator of protection against mKSA tumor development was assessed via in vivo IFN-γ neutralization, and these experiments demonstrated a requirement for this cytokine in the induction immune phase. Neutralization of IFN-γ was associated with a reduction in Th1 cytokine-producing CD4+ and CD8+ splenocytes, as assessed by flow cytometry analysis, and provided further evidence for the role of CD4+ T lymphocytes as drivers of the cellular immune response. Depletion of NK cells and CD8+ T lymphocytes demonstrated the expendability of these cell types individually, but showed a requirement for a resident cytotoxic cell population within the immune effector phase. Our findings demonstrate the importance of IFN-γ in the induction of protective immunity stimulated by pCMV-Tag DNA-based vaccine and help to clarify the general mechanisms by which DNA vaccines trigger immunity to tumor cells.


Subject(s)
Antigens, Polyomavirus Transforming/immunology , Interferon-gamma/immunology , Polyomavirus Infections/immunology , Simian virus 40/immunology , Tumor Virus Infections/immunology , Vaccines, DNA/immunology , Animals , Antigens, Polyomavirus Transforming/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cells, Cultured , Female , Interferon-gamma/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Plasmids , Polyomavirus Infections/genetics , Spleen/immunology , Th1 Cells/immunology , Tumor Virus Infections/genetics
2.
J Infect Dis ; 204(9): 1437-49, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21921206

ABSTRACT

The prophylactic efficacy of a schistosome antigen (Sm-p80) was tested in a nonhuman primate model, the baboon. Using a total of 28 baboons, different vaccination strategies were used including recombinant Sm-p80 protein formulated in Toll-like receptor 7 and Toll-like receptor 9 agonists, and DNA priming followed by boosting with protein plus adjuvants. Recombinant protein approaches provided levels of prophylactic efficacy of 52%-58%, whereas prime-boost approaches conferred 38%-47% protection in baboons. An appropriately balanced pro-inflammatory (T-helper 17 [Th17] and Th1) and anti-inflammatory (Th2) type of response was generated; the Th1 and Th17 types of immune responses appear to be indicative of increased prophylactic efficacy. Production and expression of several cytokines (interleukin 2 [IL-2], interferon γ, IL-12α, IL-1ß, IL-6, and IL-22) were up-regulated in vaccinated animals. Human correlate studies revealed Sm-p80 reactivity with immunoglobulin G in human serum samples from schistosome-infected individuals. In addition, a complete lack of prevailing Sm-p80-specific immunoglobulin E in a high-risk or infected population was observed, thus minimizing the risk of hypersensitivity reaction following vaccination with Sm-p80 in humans. This study provided the proof of concept to move Sm-p80 forward into further preclinical development leading to human clinical trials.


Subject(s)
Antibodies, Helminth/blood , Antigens, Helminth/immunology , Endemic Diseases , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/prevention & control , Vaccines, DNA/immunology , Animals , Antigens, Helminth/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Male , Papio , Primate Diseases/immunology , Primate Diseases/prevention & control , Serum/immunology , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
3.
J Virol ; 85(14): 7216-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593176

ABSTRACT

A mechanistic analysis of tumor immunity directed toward the viral oncoprotein simian virus 40 (SV40) large tumor antigen (Tag) has previously been described by our laboratory for scenarios of recombinant Tag immunization in BALB/c mice. In the present study, we performed a preliminary characterization of the immune components necessary for systemic tumor immunity induced upon immunization with plasmid DNA encoding SV40 Tag as a transgene (pCMV-Tag). Antibody responses to SV40 Tag were observed via indirect enzyme-linked immunosorbent assay following three intramuscular (i.m.) injections of pCMV-Tag and were typified by a mixed Th1/Th2 response. Complete tumor immunity within a murine model of pulmonary metastasis was achieved upon two i.m. injections of pCMV-Tag, as assessed by examination of tumor foci in mouse lungs, without a detectable antibody response to SV40 Tag. Induction-phase and effector-phase depletions of T cell subsets were performed in vivo via administration of depleting rat monoclonal antibodies, and these experiments demonstrated that CD4(+) T lymphocytes are required in both phases of the adaptive immune response. Conversely, depletion of CD8(+) T lymphocytes did not impair tumor immunity in either immune phase and resulted in the premature production of antibodies to SV40 Tag. Our findings are unique in that a dominant role could be ascribed to CD4(+) T lymphocytes within a model of DNA vaccine-induced tumor immunity to Tag-expressing tumor cells. Additionally, our findings provide insight into the general mechanisms of vaccine-induced tumor immunity directed toward tumors bearing distinct tumor-associated antigens.


Subject(s)
Antigens, Viral, Tumor/immunology , CD4-Positive T-Lymphocytes/immunology , Neoplasms, Experimental/immunology , Plasmids , Simian virus 40/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred BALB C , Vaccines, DNA/administration & dosage
4.
Clin Dev Immunol ; 2010: 697158, 2010.
Article in English | MEDLINE | ID: mdl-20936120

ABSTRACT

The employment of the immune system to treat malignant disease represents an active area of biomedical research. The specificity of the immune response and potential for establishing long-term tumor immunity compels researchers to continue investigations into immunotherapeutic approaches for cancer. A number of immunotherapeutic strategies have arisen for the treatment of malignant disease, including various vaccination schemes, cytokine therapy, adoptive cellular therapy, and monoclonal antibody therapy. This paper describes each of these strategies and discusses some of the associated successes and limitations. Emphasis is placed on the integration of techniques to promote optimal scenarios for eliminating cancer.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cytokines/administration & dosage , Cytokines/immunology , Dendritic Cells/immunology , Dogs , Humans , Mice
5.
J Virol ; 84(19): 10121-30, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20668083

ABSTRACT

We examined properties of the innate immune response against the tumor-specific antigen simian virus 40 (SV40) large tumor antigen (Tag) following experimental pulmonary metastasis in naive mice. Approximately 14 days after mKSA tumor cell challenge, expression of inflammatory mediators such as tumor necrosis factor alpha (TNF-alpha), interleukin-2 (IL-2), and RANTES was upregulated in splenocytes harvested from mice, as assessed by flow cytometry and antibody array assays. This response was hypothesized to activate and induce tumor-directed NK cell lysis since IL-2-stimulated NK cells mediated tumor cell destruction in vitro. The necessary function of NK cells was further validated in vivo through selected antibody depletion of NK cells, which resulted in an overwhelming lung tumor burden relative to that in animals receiving a control rabbit IgG depletion regimen. Interestingly, mice achieved increased protection from experimental pulmonary metastasis when NK cells were further activated indirectly through in vivo administration of poly(I:C), a Toll-like receptor 3 (TLR3) agonist. In a separate study, mice receiving treatments of poly(I:C) and recombinant SV40 Tag protein immunization mounted effective tumor immunity in an established experimental pulmonary metastasis setting. Initiating broad-based immunity with poly(I:C) was observed to induce a Th1 bias in the SV40 Tag antibody response that led to successful antitumor responses not observed in animals treated only with poly(I:C) or SV40 Tag. These data have direct implications for immunotherapeutic strategies incorporating methods to elicit inflammatory reactions, particularly NK cell-driven lysis, against malignant cell types that express a tumor-specific antigen such as SV40 Tag.


Subject(s)
Antigens, Polyomavirus Transforming/immunology , Immunity, Innate , Lung Neoplasms/immunology , Animals , Antibodies, Neoplasm/biosynthesis , Cell Line, Tumor , Cytotoxicity, Immunologic , Immunotherapy , In Vitro Techniques , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Poly I-C/pharmacology , Rabbits , Th1 Cells/immunology , Toll-Like Receptors/metabolism
6.
J Infect Dis ; 201(7): 1105-12, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20187746

ABSTRACT

To date, no vaccine is available to prevent human schistosomiasis. We have targeted a protein of Schistosoma mansoni that plays an important role in the surface membrane renewal process, a mechanism widely believed to be utilized by the parasite as an immune evasion strategy. Sm-p80 antigen is a promising vaccine target because of its documented immunogenicity, protective efficacy, and antifecundity effects observed in both experimental murine and nonhuman primate models of this infectious disease. In the present study, we report that, in a vector approved for human use (VR1020), an Sm-p80-based DNA vaccine formulation confers a 46% reduction in the worm burden in a baboon (Papio anubis) model. Baboons vaccinated with Sm-p80-VR1020 had a 28% decrease in egg production after challenge with the infectious parasite. Sm-p80-VR1020 vaccine elicited robust immune responses to specific antigen Sm-p80, including immunoglobulin (Ig) G, its subtypes IgG1 and IgG2, and IgA and IgM in vaccinated animals. When stimulated in vitro with recombinant Sm-p80, peripheral blood mononuclear cells and splenocytes from baboons vaccinated with Sm-p80-VR1020 produced considerably higher levels of T helper 1 response-enhancing cytokines (interleukin [IL]-2 and interferon-gamma) than T helper 2 (Th2) response-enhancing cytokines (IL-4 and IL-10). Peripheral blood mononuclear cells produced a significantly higher number of spot-forming units for interferon-gamma than for IL-4 in enzyme-linked immunosorbent spot assays. A mixed T helper 1/T helper 2 type of humoral and T cell responses was generated after immunization with Sm-p80-VR1020. These findings again highlight the potential of Sm-p80 as a promising vaccine candidate for schistosomiasis.


Subject(s)
Antigens, Helminth/immunology , Papio/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Vaccines, DNA/immunology , Animals , Antibodies, Helminth/blood , Antigens, Helminth/genetics , CHO Cells , COS Cells , Cell Proliferation , Chlorocebus aethiops , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Helminth Proteins/genetics , Helminth Proteins/immunology , Immunoglobulin G/blood , Interleukin-4/metabolism , Intestines/parasitology , Leukocytes, Mononuclear/metabolism , Liver/parasitology , Parasite Egg Count , Schistosoma mansoni/genetics , Schistosomiasis mansoni/immunology , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage
7.
J Virol ; 84(2): 883-93, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889780

ABSTRACT

The required activities of CD4(+) T cells and antibody against the virally encoded oncoprotein simian virus 40 (SV40) Tag have previously been demonstrated by our laboratory to be mediators in achieving antitumor responses and tumor protection through antibody-dependent cell-mediated cytotoxicity (ADCC). In this study, we further characterize the necessary immune cell components that lead to systemic tumor immunity within an experimental pulmonary metastatic model as the result of SV40 Tag immunization and antibody production. Immunized animals depleted of CD8(+) T cells at the onset of experimental tumor cell challenge developed lung tumor foci and had an overall decreased survival due to lung tumor burden, suggesting a role for CD8(+) T cells in the effector phase of the immune response. Lymphocytes and splenocytes harvested from SV40 Tag-immunized mice experimentally inoculated with tumor cells synthesized increased in vitro levels of the Th1 cytokine gamma interferon (IFN-gamma), as assessed by enzyme-linked immunosorbent assay (ELISA) and flow cytometry assays. CD8(+) T-cell activity was also heightened in SV40 Tag-immunized and tumor cell-challenged mice, based upon intracellular production of perforin, confirming the cytolytic properties of CD8(+) T cells against tumor cell challenge. Altogether, these data point to the role of recombinant SV40 Tag protein immunization in initiating a cytotoxic T-lymphocyte (CTL) response during tumor cell dissemination and growth. The downstream activity of CD8(+) T cells within this model is likely initiated from SV40 Tag-specific antibody mediating ADCC tumor cell destruction.


Subject(s)
Antigens, Polyomavirus Transforming/immunology , CD8-Positive T-Lymphocytes/immunology , Lung Neoplasms/immunology , Simian virus 40/immunology , Tumor Virus Infections/immunology , Animals , Antibodies, Viral/blood , Antigens, Polyomavirus Transforming/administration & dosage , Cell Line, Transformed , Immunity, Humoral , Immunization , Kidney/cytology , Kidney/virology , Lung Neoplasms/mortality , Lung Neoplasms/prevention & control , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic , Th1 Cells/immunology , Tumor Virus Infections/mortality , Tumor Virus Infections/prevention & control
8.
Vaccine ; 27(21): 2830-7, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19366570

ABSTRACT

Schistosomiasis is an important parasitic disease for which there is no available vaccine. We have focused on a functionally important antigen of Schistosoma mansoni, Sm-p80, as a vaccine candidate because of its consistent immunogenicity, protective potential and antifecundity effect observed in murine models; and for its pivotal role in the immune evasion process. In the present study we report that an Sm-p80-based DNA vaccine formulation confers 38% reduction in worm burden in a nonhuman primate model, the baboon (Papio anubis). Animals immunized with Sm-p80-pcDNA3 exhibited a decrease in egg production by 32%. Sm-p80 DNA elicited specific immune responses that include IgG; its subtypes IgG1 and IgG2; and IgM in vaccinated animals. Peripheral blood mononuclear cells (PBMCs) from immunized animals when stimulated in vitro with Sm-p80 produced appreciably more Th1 response enhancing cytokines (IL-2, IFN-gamma) than Th2 response enhancing cytokines (IL-4, IL-10). PBMCs produced appreciably more spot-forming units for INF-gamma than for IL-4 in enzyme-linked immunosorbent spot (ELISPOT) assays. Overall it appears that even though a mixed (Th1/Th2) type of humoral antibody response was generated following immunization with Sm-p80; the dominant protective immune response is Th1 type. These data reinforce the potential of Sm-p80 as an excellent vaccine candidate for schistosomiasis.


Subject(s)
Antigens, Helminth/immunology , Antigens, Helminth/metabolism , Fertility , Papio/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Helminth/genetics , Cell Line , Chlorocebus aethiops , Cricetinae , Cytokines/biosynthesis , Cytokines/immunology , Female , Male , Models, Animal , Ovum/immunology , Schistosoma mansoni/genetics , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/immunology , T-Lymphocytes/immunology , Vaccines, DNA/genetics
9.
J Immunother ; 31(8): 693-703, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18779750

ABSTRACT

For ovarian cancer (OC) patients with advanced or metastatic disease, standard treatments (chemotherapy and radiotherapy) are not very effective and have undesirable side effects. Newer and more promising approaches in cancer treatment use components of the immune system. In this study, we applied an adoptive immunotherapy-based approach using a cancer testis antigen, sperm protein 17, as a target for the treatment of human metastatic OC in a NOD.CB17-PrkDCcid/J (nonobese, diabetic severe combined immunodeficient) mouse model. We used the human SK-OV-3A2.A3 OC cell line, endogenously expressing sperm protein 17, to induce tumor growth in mice. We provide direct evidence, for the first time, that in vitro cultured, monoclonal, cytotoxic T lymphocytes (derived either from advanced OC patients or from healthy donors), specific for sperm protein 17, can eradicate human metastatic OC cells. In addition, we observed no evidence of autoimmunity after histologic examination of the tissue sections adding to the safety profile of our approach.


Subject(s)
Antigens, Surface/immunology , Carrier Proteins/immunology , Immunotherapy, Adoptive/methods , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , T-Lymphocytes, Cytotoxic/transplantation , Animals , Antigens, Surface/metabolism , Calmodulin-Binding Proteins , Carrier Proteins/metabolism , Cell Line, Tumor , Female , Humans , Male , Membrane Proteins , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology
10.
Virology ; 377(1): 54-62, 2008 Jul 20.
Article in English | MEDLINE | ID: mdl-18485439

ABSTRACT

Simian virus 40 (SV40) is a polyomavirus for which non-human primates are the permissive host. The baboon (Papio spp.) is an old world monkey that is used in a variety of research investigations; however, natural infection of SV40 among baboons has not been thoroughly examined or reported. Initially, we were interested in determining the prevalence of SV40 infection among a captive colony of baboons based on the presence of antibodies to SV40 large T-antigen (Tag). An overall seroprevalence rate of >50% was found after screening sera from 142 baboons in the colony based on ELISA. Endpoint titer values for serum antibody binding to SV40 Tag reached as high as 1280 for 5 out of 142 baboons. Peptide binding assays revealed that a range of SV40 Tag epitopes are immunogenic in the baboon, and that individual animals differ in their humoral immune responses to SV40 Tag based on epitope recognition. Specificity to SV40 Tag and not some other primate polyomavirus encoded large Tag was further examined by serologic reactivity to peptide epitopes unique to SV40 Tag. Additional serology was performed to assess SV40 Tag reactivity by Western blot and whether antibodies were capable of neutralizing SV40 infectivity in vitro. Although antibodies with high levels of SV40 neutralization were observed in a number of the baboons, there was a lack of correlation between viral neutralization and antibodies to SV40 Tag. Further examination using molecular-based diagnosis and SV40 Tag specific real-time quantitative PCR determined that some of the baboons appeared to be exposed to SV40. DNA sequence analysis of the PCR products confirmed that SV40 Tag specific sequences were detected in baboons.


Subject(s)
Papio/virology , Simian virus 40/isolation & purification , Amino Acid Sequence , Animals , Animals, Laboratory/immunology , Animals, Laboratory/virology , Antibodies, Viral/blood , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/isolation & purification , Base Sequence , DNA Primers/genetics , DNA, Viral/genetics , Enzyme-Linked Immunosorbent Assay , Female , Male , Molecular Sequence Data , Monkey Diseases/immunology , Monkey Diseases/virology , Papio/immunology , Papio anubis/immunology , Papio anubis/virology , Papio cynocephalus/immunology , Papio cynocephalus/virology , Papio ursinus/immunology , Papio ursinus/virology , Polymerase Chain Reaction , Polyomavirus Infections/immunology , Polyomavirus Infections/veterinary , Polyomavirus Infections/virology , Sequence Homology, Nucleic Acid , Seroepidemiologic Studies , Simian virus 40/genetics , Simian virus 40/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/veterinary , Tumor Virus Infections/virology
11.
Parasitol Res ; 102(5): 825-33, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18259777

ABSTRACT

Considerable morbidity and mortality results from the affliction of an estimated 200 million people worldwide by several species of schistosomes; 779 million are exposed to the disease in 74 different countries. Even though anti-parasitic drugs and other control measures, including public hygiene and snail control are available, the advent of an effective vaccine still remains the most potentially powerful means for the control of this disease. The putative vaccine could be administered to small children prior to the time when their contact with infected water is maximal, so as to prevent severe infection in the subsequent years. This review attempts to summarize the status of schistosome vaccine development with special emphasis on functionally important vaccine candidates. The importance of utilizing both murine and nonhuman primate models as a prerequisite for clinical trials is discussed.


Subject(s)
Disease Models, Animal , Schistosomiasis/prevention & control , Vaccines , Animals , Antigens, Helminth/immunology , Child , Child, Preschool , Humans , Mice , Papio , Schistosomiasis/parasitology , Vaccines/administration & dosage , Vaccines/immunology
12.
J Immunol Methods ; 321(1-2): 86-93, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17336323

ABSTRACT

No experimental animal model employing a primary human ovarian carcinoma (OC) cell line is presently available that tracks the progression of this cell line with an identifiable marker. This hinders investigations related to developing new approaches for treating OC. Here, we describe the development of a tumor model in NOD/SCID mice for human OC that makes use of the endogenously expressed tumor specific sperm protein 17 (Sp17) cancer testis antigen. In this model, human SKOV-3 OC cell lines were intra-peritoneally seeded. Subsequently viable SKOV-3 cells were recovered from primary organ cell cultures from the liver ovaries, abdomen, and ascitic fluid, and their presence was confirmed by the detection of Sp17 mRNA by RT-PCR and Sp17 protein by immunocytochemistry and FACS analysis. When SKOV-3 tumor cells were administered intravenously the mice developed primarily lung tumor foci. This model makes it possible to evaluate new immunotherapeutic strategies for the treatment of human OC based on the biomarker Sp17.


Subject(s)
Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Neoplasms, Experimental/metabolism , Ovarian Neoplasms/metabolism , Animals , Antigens, Surface/genetics , Biomarkers, Tumor/genetics , Calmodulin-Binding Proteins , Carrier Proteins/genetics , Cell Line, Tumor , Cell Separation , Disease Progression , Female , Flow Cytometry , Humans , Immunohistochemistry , Injections, Intraperitoneal , Injections, Intravenous , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Membrane Proteins , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation/methods , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
13.
Int J Cancer ; 121(1): 88-94, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17290395

ABSTRACT

The aim of this study is to investigate whether an active immunoprophylactic approach combining specific antigens and adjuvant stimuli would be able to inhibit prostate carcinogenesis in transgenic TRAMP mice. A vaccine consisting of allogeneic large T antigen (TAg)-positive SV40-transformed cells combined with systemic recombinant IL-12 was administered to TRAMP mice, starting from when they were still tumor-free at 5-6 weeks of age. The combined vaccine significantly inhibited prostate carcinogenesis, giving a more than doubled median latency time of prostatic tumors (53 weeks in comparison to 26 weeks in control mice). Vaccination with cells alone or IL-12 treatment alone was poorly effective (median latency of 30 and 39 weeks, respectively). The combined vaccine induced a very high CD4 response biased toward the Th1 pathway, with the induction of a humoral response that included TAg-specific antibodies. Therefore, such active immunoprophylactic approach based on the combination of allogeneic SV40 TAg-positive cells and systemic administration of recombinant IL-12 significantly delayed autochthonous urogenital carcinogenesis driven by SV40 TAg in TRAMP mice.


Subject(s)
Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/prevention & control , Animals , Antibody Formation/immunology , Cancer Vaccines , Cell Line , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Survival Rate
14.
J Virol ; 81(3): 1313-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17108042

ABSTRACT

Simian virus 40 (SV40) large tumor antigen (Tag) represents a virus-encoded tumor-specific antigen expressed in many types of human cancers and a potential immunologic target for antitumor responses. Fc receptors are important mediators in the regulation and execution of host effector mechanisms against conditions including infectious diseases, autoimmunity, and cancer. By examining tumor protection in SV40 Tag-immunized wild-type BALB/c mice using an experimental pulmonary metastasis model, we attempted to address whether engagement of the immunoglobulin G Fc receptors (FcgammaRs) on effector cells is necessary to mediate antitumor responses. All immunized BALB/c FcgammaR-/- knockout mice developed anti-SV40 Tag antibody responses prior to experimental challenge with a tumorigenic cell line expressing SV40 Tag. However, all mice deficient in the activating FcgammaRI (CD64) and FcgammaRIII (CD16) were unable to mount protective immunologic responses against tumor challenge and developed tumor lung foci. In contrast, mice lacking the inhibitory receptor FcgammaRII (CD32) demonstrated resistance to tumorigenesis. These results underscore the importance of effector cell populations expressing FcgammaRI/III within this murine tumor model system, and along with the production of a specific humoral immune response, antibody-dependent cell-mediated cytotoxicity (ADCC) may be a functioning mechanism of tumor clearance. Additionally, these data demonstrate the potential utility of ADCC as a viable approach for targeting vaccination strategies that promote FcgammaRI/III scavenging pathways against cancer.


Subject(s)
Antigens, Polyomavirus Transforming/immunology , Immunity, Cellular , Lung Neoplasms/prevention & control , Receptors, IgG/physiology , Animals , Cell Transformation, Viral/immunology , Fibroblasts/virology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Simian virus 40/immunology
15.
J Cell Biochem ; 98(2): 235-42, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16440328

ABSTRACT

Vaccination with plasmid DNA is an active area of investigation that is being applied to diseases including cancer and microbial pathogens associated with infectious diseases. Since its discovery, great progress has been made with the administration of DNA vaccines to initiate specific and effective immune responses against targeted illnesses. However, many obstacles still face its use in prophylactic and therapeutic vaccination scenarios. The nature of these difficulties alongside the successes and future of plasmid DNA will be discussed.


Subject(s)
Communicable Disease Control/methods , Neoplasms/prevention & control , Vaccines, DNA/therapeutic use , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/therapeutic use , Disease Models, Animal , History, 20th Century , Humans , Immunization , Immunotherapy, Active/methods , Neoplasms/immunology , Plasmids/genetics , Plasmids/immunology , Vaccination/history , Vaccination/methods , Vaccines, DNA/immunology
17.
Cancer Res ; 65(7): 2938-46, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805297

ABSTRACT

The nonimmunogenic 4T1 murine mammary carcinoma model and a model surrogate tumor antigen (sTA) were employed to explore the possibility of inducing tumor-specific immunity through active immunization in the absence of defined tumor-associated antigens. Immunization of naive mice with protein-based sTA resulted in protection from s.c. challenge, with 4T1 modified to express the sTA (4T1.sTA), or from a sTA-expressing unrelated tumor cell line (mKSA). Immunization had no effect on parental 4T1 tumor growth or the formation of parental 4T1 spontaneous lung metastases. Mice that were sTA immunized and successfully rejected 4T1.sTA challenge also rejected a subsequent challenge in the contralateral flank with parental 4T1 and strikingly prevented the formation of spontaneous parental 4T1 lung metastases. The rejection of parental 4T1 seemed to be specific for and associated with unknown 4T1 tumor-associated antigens, because rejection of mKSA did not induce cross-protection against a challenge with parental 4T1. To evaluate the effect of this vaccine approach on established disease, mice were simultaneously challenged on day 0 with 4T1.sTA and parental 4T1 in contralateral flanks and then immunized on days 3, 10, 17, and 24 with sTA protein. Tumor growth and metastasis were delayed in four of five animals, and 20% (2 of 5) of the animals were tumor free at the completion of the experiment. Together, these data suggest that prior vaccination with a sTA followed by inoculation with poorly immunogenic tumor cells modified to express the sTA activates determinant spreading and the induction of systemic tumor immunity resulting in indigenous tumor rejection.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Polyomavirus Transforming/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Animals , Cell Line, Tumor , Female , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , T-Lymphocytes/immunology
18.
Vaccine ; 23(12): 1451-6, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15670880

ABSTRACT

Even though schistosomicidal agents and other control measures, including public hygiene and snail control exist, development of an efficacious vaccine still remains the most potentially powerful method for control of schistosomiasis. In our continuing efforts to develop a vaccine against schistosomiasis, we have selected a vaccine candidate (Sm-p80), which plays an important role in the immune evasion process of the parasite. Sm-p80 has been shown to confer up to 60% protection in mice following experimental infection. In this initial study, we have used Sm-p80 plus the Th1 response promoting cytokine, interleukin-2 (IL-2), in a DNA immunogen formulation. The vaccine was tested for its safety and immunogenicity in a baboon model of schistosomiasis. The vaccine generated a Th1 type Sm-p80-specific response in baboons with IgG(1)/IgG(2) ratios of less than 1.0. No detectable IgG(3) or IgG(4) anti-Sm-p80 responses were present in the immunized baboons. The antibodies to Sm-p80 were able to kill up to 35% schistosomula in vitro in the presence of complement. These results although preliminary suggest the potential of Sm-p80 as a viable vaccine candidate for schistosomiasis.


Subject(s)
Antigens, Helminth/immunology , Schistosoma/immunology , Schistosomiasis/immunology , Schistosomiasis/prevention & control , Vaccines, DNA/immunology , Animals , Antibodies, Helminth/blood , Antigens, Helminth/genetics , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/blood , Interleukin-2/genetics , Interleukin-2/immunology , Models, Animal , Papio , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
19.
Virology ; 332(1): 28-37, 2005 Feb 05.
Article in English | MEDLINE | ID: mdl-15661138

ABSTRACT

Simian virus 40 (SV40) contains an essential protein, large tumor antigen (Tag), which assists in viral replication and causes cell transformation and immortalization. Our laboratory has examined plasmid DNA, expressing SV40 Tag under two different promoters, for use in potential cancer vaccination strategies. One plasmid, pSV3-neo, failed to induce SV40 Tag antibody, produced a weak cell-mediated response, and only partial protection in murine experimental tumor challenge systems. The second plasmid, pCMV-Tag, induced antibodies to SV40 Tag, produced a robust cell-mediated response, and invoked complete tumor immunity in vivo. The induction of CD4+ and CD8+ T cell responses following plasmid DNA immunization and tumor cell challenge reflected a type 1 cytokine secretion profile. Our hypothesis for this differential immune response is that pCMV-Tag exhibits a higher level of transgene expression due to a more efficient promoter. We determined that pCMV-Tag levels of SV40 Tag mRNA and protein expression were higher when compared to pSV3-neo. A threshold amount of SV40 Tag may be required to stimulate antibody production and provide complete systemic tumor immunity.


Subject(s)
Antigens, Polyomavirus Transforming/immunology , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Simian virus 40/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Transformed , Immunization , Mice , Mice, Inbred BALB C , Sarcoma, Experimental/immunology
20.
Crit Rev Immunol ; 24(2): 129-56, 2004.
Article in English | MEDLINE | ID: mdl-15581394

ABSTRACT

Over the last decade, a number of concerns have arisen related to safety issues that have had an adverse effect on the public's trust, particularly among parents whose children are the primary recipient of the vaccine. Historically, the live attenuated measles virus (MV) vaccine and the combination multivalent measles, mumps, and rubella (MMR) vaccine have had a major impact on the health of children worldwide and have been extremely successful at preventing infectious diseases associated with three childhood viral pathogens. In this report, we describe MV infection, replication, pathogenesis, and immunization. MV is a viral pathogen that exhibits a number of complex processes that can effect its replication, pathogenesis, and the induction of an effective antiviral immune response. We describe the published literature as it relates to MV infection and immunization and report adverse events in an attempt to provide a balanced discussion and an historical perspective of the MMR vaccine and autism.


Subject(s)
Autistic Disorder/etiology , Measles virus/pathogenicity , Measles-Mumps-Rubella Vaccine/adverse effects , Measles/prevention & control , Contraindications , Humans , Immunity, Active , Measles virus/immunology , Measles virus/physiology , Measles-Mumps-Rubella Vaccine/therapeutic use , Models, Biological , T-Lymphocytes/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...