Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Immunol Rev ; 222: 43-69, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18363994

RESUMO

We contrast the efforts to treat ovarian cancer and cervical cancer through vaccination because of their different pathobiology. A plethora of approaches have been developed for therapeutic vaccination against cancer, many of which target defined tumor-associated antigens (TAAs). Persistent infection with oncogenic human papillomavirus (HPV) types causes cervical cancer. Furthermore, cervical cancer patients frequently mount both humoral and T-cell immune responses to the HPV E6 and E7 oncoproteins, whose expression is required for the transformed phenotype. Numerous vaccine studies target these viral TAAs, including recent trials that may enhance clearance of pre-malignant disease. By contrast, little is known about the etiology of epithelial ovarian cancer. Although it is clear that p53 mutation or loss is a critical early event in the development of epithelial ovarian cancer, no precursor lesion has been described for the most common serous histotype, and even the location of its origin is debated. These issues have complicated the selection of appropriate ovarian TAAs and the design of vaccines. Here we focus on mesothelin as a promising ovarian TAA, because it is overexpressed and immunogenic at high frequency in patients, is displayed on the cell surface, and potentially contributes to ovarian cancer biology.


Assuntos
Antígenos Virais de Tumores/imunologia , Biomarcadores Tumorais , Vacinas Anticâncer , Epitopos/imunologia , Imunoterapia Ativa/métodos , Glicoproteínas de Membrana/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias do Colo do Útero/imunologia , Autoanticorpos/imunologia , Células Dendríticas , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Proteínas Ligadas por GPI , Vetores Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Linfócitos do Interstício Tumoral , Mesotelina , Regressão Neoplásica Espontânea , Estadiamento de Neoplasias , Proteínas Oncogênicas Virais/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Proteínas E7 de Papillomavirus/imunologia , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/imunologia , Valor Preditivo dos Testes , Proteínas/imunologia , Linfócitos T Reguladores , Proteína Supressora de Tumor p53/imunologia , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/terapia
2.
Cancer Immunol Immunother ; 60(8): 1085-96, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21526359

RESUMO

Ovarian cancer is currently the most lethal gynecologic cancer in the United States. There is an urgent need for the development of innovative therapies against ovarian cancer, such as immunotherapy. The toll-like receptor 3 ligand, polyriboinosinic:polyribocytidylic acid (poly(I:C), has emerged as a promising adjuvant for activating the host immune responses for the control of tumors. We reasoned that a strategy to enhance the intracellular uptake of poly(I:C) will likely improve the poly(I:C) adjuvant effect. Since polyethylenimine (PEI) has been shown to increase the transfection efficiency of nucleic acids, we characterized the antitumor effects in mouse ovarian surface epithelial cells (MOSEC) tumor-bearing mice treated intraperitoneally with poly(I:C) and PEI. We observed that tumor-bearing mice treated with poly(I:C) and PEI generated significantly better therapeutic antitumor effects against MOSEC tumors compared with treatment with poly(I:C) alone. Furthermore, we found that NK cells play a significant role in the antitumor effects generated by treatment with poly(I:C) in combination with PEI. Intraperitoneal administration of poly(I:C) with PEI led to the uptake of poly(I:C) mainly by CD11b+ macrophages, resulting in the high expression of MHC class II and IL-12 (M1 phenotype). In addition, adoptive transfer of CD11b+ macrophages from mice treated with poly(I:C) and PEI was found to lead to increased number of activated NK cells in the recipient mice. Taken together, our data indicate that PEI can potentially be used to improve the uptake of poly(I:C) by CD11b+ macrophages, leading to the activation of NK cells and the control of murine ovarian tumors.


Assuntos
Carcinoma/terapia , Imunoterapia , Macrófagos/efeitos dos fármacos , Neoplasias Ovarianas/terapia , Polietilenoimina/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígeno CD11b/biossíntese , Carcinoma/imunologia , Carcinoma/patologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Imunidade Inata/efeitos dos fármacos , Injeções Intraperitoneais , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Poli I-C/administração & dosagem
3.
J Biomed Sci ; 18: 21, 2011 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21385449

RESUMO

Antigen-specific immunotherapy using DNA vaccines has emerged as an attractive approach for the control of tumors. Another novel cancer therapy involves the employment of the vascular disrupting agent, 5,6-dimethylxanthenone-4-acetic acid (DMXAA). In the current study, we aimed to test the combination of DMXAA treatment with human papillomavirus type 16 (HPV-16) E7 DNA vaccination to enhance the antitumor effects and E7-specific CD8+ T cell immune responses in treated mice. We determined that treatment with DMXAA generates significant therapeutic effects against TC-1 tumors but does not enhance the antigen-specific immune responses in tumor bearing mice. We then found that combination of DMXAA treatment with E7 DNA vaccination generates potent antitumor effects and E7-specific CD8+ T cell immune responses in the splenocytes of tumor bearing mice. Furthermore, the DMXAA-mediated enhancement or suppression of E7-specific CD8+ T cell immune responses generated by CRT/E7 DNA vaccination was found to be dependent on the time of administration of DMXAA and was also applicable to other antigen-specific vaccines. In addition, we determined that inducible nitric oxide synthase (iNOS) plays a role in the immune suppression caused by DMXAA administration before DNA vaccination. Our study has significant implications for future clinical translation.


Assuntos
Antineoplásicos/uso terapêutico , Vacinas Anticâncer/administração & dosagem , Neoplasias Experimentais/terapia , Vacinas contra Papillomavirus/administração & dosagem , Xantonas/administração & dosagem , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neoplasias Experimentais/imunologia , Vacinas contra Papillomavirus/genética , Vacinas contra Papillomavirus/uso terapêutico , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/uso terapêutico
4.
Mol Ther ; 18(4): 692-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20087318

RESUMO

Cancer therapy using oncolytic viruses represents a promising new approach for controlling ovarian cancer. In this study, we have circumvented the limitation of repeated vaccination by employing different virus vectors, Semliki Forest Virus (SFV) and vaccinia virus (VV) for boosting the immune response. We found that infection of tumor-bearing mice with VV followed by infection with SFV or vice versa leads to enhanced antitumor effects against murine ovarian surface epithelial carcinoma (MOSEC) tumors. Furthermore, infection with VV-ovalbumin (OVA) followed by infection with SFV-OVA or vice versa was found to lead to enhanced OVA-specific CD8(+) T-cell immune responses. In addition, we found that infection with SFV-OVA followed by infection with VV-OVA leads to enhanced antitumor effects in vivo and enhanced tumor killing in vitro through a combination of viral oncolysis and antigen-specific immunity. The clinical implications of this study are discussed.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia , Terapia Viral Oncolítica , Neoplasias Ovarianas/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Terapia Genética/métodos , Vetores Genéticos , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/imunologia , Vírus da Floresta de Semliki/imunologia , Vaccinia virus/imunologia
5.
Eur J Immunol ; 39(2): 491-506, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19180469

RESUMO

IL-15 and IL-15 receptor alpha (IL-15RA) play a significant role in multiple aspects of T-cell biology. However, given the evidence that IL-15RA can present IL-15 in trans, the functional capacity of IL-15RA expressed on CD8(+) T cells to modify IL-15 functions in cis is currently unclear. In the current study, we explore the functional consequences of IL-15RA, expression on T cells using a novel method to transfect naive CD8(+) T cells. We observed that RNA nucleofection led to highly efficient, non-toxic, and rapid manipulation of protein expression levels in unstimulated CD8(+) T cells. We found that transfection of unstimulated CD8(+) T cells with IL-15RA RNA led to enhanced viability of CD8(+) T cells in response to IL-15. Transfection with IL-15RA enhanced IL-15-mediated phosphorylation of STAT5 and also promoted IL-15-mediated proliferation in vivo of adoptively transferred naïve CD8(+) T cells. We demonstrated that IL-15RA can present IL-15 via cis-presentation on CD8(+) T cells. Finally, we showed that transfection with a chimeric construct linking IL-15 to IL-15RA cell autonomously enhances the viability and proliferation of primary CD8(+) T cells and cytotoxic potential of antigen-specific CD8(+) T cells. The clinical implications of the current study are discussed.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Subunidade alfa de Receptor de Interleucina-15/biossíntese , Interleucina-15/biossíntese , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/transplante , Proliferação de Células , Sobrevivência Celular/imunologia , Interferon gama/imunologia , Interleucina-15/genética , Subunidade alfa de Receptor de Interleucina-15/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Fator de Transcrição STAT5/metabolismo , Transfecção
6.
J Biomed Sci ; 17: 32, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20426849

RESUMO

BACKGROUND: There is an urgent need to develop new innovative therapies for the control of advanced cancer. The combination of antigen-specific immunotherapy with the employment of immunomodulatory agents has emerged as a potentially plausible approach for the control of advanced cancer. METHODS: In the current study, we explored the combination of the DNA vaccine encoding calreticulin (CRT) linked to human papillomavirus type 16 (HPV-16) E7 antigen (CRT/E7) with the TLR7 agonist imiquimod for their ability to generate E7-specific immune responses and antitumor effects in tumor-bearing mice. RESULTS: We observed that treatment with CRT/E7 DNA in combination with imiquimod leads to an enhancement in the E7-specific CD8+ T cell immune responses and a decrease in the number of myeloid-derived suppressor cells in the tumor microenvironment of tumor-bearing mice. Furthermore, treatment with CRT/E7 DNA in combination with imiquimod leads to significantly improved antitumor effects and prolonged survival in treated mice. In addition, treatment with imiquimod led to increased number of NK1.1+ cells and F4/80+ cells in the tumor microenvironment. Macrophages and NK1.1+ cells were found to play an important role in the antitumor effects mediated by treatment with CRT/E7 DNA in combination with imiquimod. CONCLUSIONS: Thus, our data suggests that the combination of therapeutic HPV DNA vaccination with topical treatment with the TLR7 agonist imiquimod enhances the antitumor immunity induced by DNA vaccination. The current study has significant implications for future clinical translation.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Aminoquinolinas/administração & dosagem , Neoplasias Experimentais/terapia , Vacinas contra Papillomavirus/administração & dosagem , Animais , Linfócitos T CD8-Positivos/imunologia , Calreticulina/genética , Calreticulina/imunologia , Linhagem Celular Tumoral , Feminino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/imunologia , Humanos , Imiquimode , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/imunologia , Vacinas contra Papillomavirus/genética , Linfócitos T Reguladores/imunologia , Receptor 7 Toll-Like/agonistas , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
7.
J Biomed Sci ; 17: 88, 2010 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-21092195

RESUMO

BACKGROUND: Effective vaccination against human papillomavirus (HPV) represents an opportunity to control cervical cancer. Peptide-based vaccines targeting HPV E6 and/or E7 antigens while safe, will most likely require additional strategies to enhance the vaccine potency. METHODS: We tested the HPV-16 E7 peptide-based vaccine in combination with a strategy to enhance CD4+ T help using a Pan HLA-DR epitope (PADRE) peptide and a strategy to enhance dendritic cell activation using the toll-like receptor 3 ligand, poly(I:C). RESULTS: We observed that mice vaccinated with E7 peptide-based vaccine in combination with PADRE peptide and poly(I:C) generated better E7-specific CD8+ T cell immune responses as well as significantly improved therapeutic anti-tumor effects against TC-1 tumors compared to E7 peptide-based vaccine with either PADRE peptide or poly(I:C) alone. Furthermore, we found that intratumoral vaccination with the E7 peptide in conjunction with PADRE peptide and poly(I:C) generates a significantly higher frequency of E7-specific CD8+ T cells as well as better survival compared to subcutaneous vaccination with the same regimen in treated mice. CONCLUSIONS: The combination of PADRE peptide and poly(I:C) with antigenic peptide is capable of generating potent antigen-specific CD8+ T cell immune responses and antitumor effects in vaccinated mice. Our study has significant clinical implications for peptide-based vaccination.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Proteínas E7 de Papillomavirus/imunologia , Vacinas contra Papillomavirus/imunologia , Peptídeos/imunologia , Animais , Antivirais/imunologia , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Papillomaviridae/imunologia , Proteínas E7 de Papillomavirus/genética , Peptídeos/genética , Poli I-C/imunologia , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/prevenção & controle , Neoplasias do Colo do Útero/virologia
8.
J Biomed Sci ; 17: 61, 2010 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-20659328

RESUMO

BACKGROUND: Ovarian cancer is the leading cause of death among women with gynecologic malignancies in the United States. Advanced ovarian cancers are difficult to cure with the current available chemotherapy, which has many associated systemic side effects. Doxorubicin is one such chemotherapeutic agent that can cause cardiotoxicity. Novel methods of delivering chemotherapy without significant side effects are therefore of critical need. METHODS: In the current study, we generated an irradiated tumor cell-based drug delivery system which uses irradiated tumor cells loaded with the chemotherapeutic drug, doxorubicin. RESULTS: We showed that incubation of murine ovarian cancer cells (MOSEC) with doxorubicin led to the intracellular uptake of the drug (MOSEC-dox cells) and the eventual death of the tumor cell. We then showed that doxorubicin loaded MOSEC-dox cells were able to deliver doxorubicin to MOSEC cells in vivo. Further characterization of the doxorubicin transfer revealed the involvement of cell contact. The irradiated form of the MOSEC-dox cells were capable of treating luciferase-expressing MOSEC tumor cells (MOSEC/luc) in C57BL/6 mice as well as in athymic nude mice resulting in improved survival compared to the non drug-loaded irradiated MOSEC cells. Furthermore, we showed that irradiated MOSEC-dox cells was more effective compared to an equivalent dose of doxorubicin in treating MOSEC/luc tumor-bearing mice. CONCLUSIONS: Thus, the employment of drug-loaded irradiated tumor cells represents a potentially innovative approach for the delivery of chemotherapeutic drugs for the control of ovarian tumors.


Assuntos
Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Tratamento Farmacológico/métodos , Neoplasias Ovarianas/tratamento farmacológico , Animais , Doxorrubicina/metabolismo , Doxorrubicina/farmacocinética , Feminino , Citometria de Fluxo , Luciferases , Camundongos , Camundongos Endogâmicos C57BL , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/efeitos da radiação
9.
J Immunol ; 181(12): 8237-47, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19050240

RESUMO

IL-15 is an important cytokine involved in the survival and function of CD8(+) T cells and NK cells. IL-15 can be presented by IL-15Ralpha (IL-15RA) to bind with the shared IL-2/IL-15Rbeta and common gamma-chains, which activate signaling pathways on NK cells and CD8(+) T cells. In the present study, we characterized the function of trans-presented IL-15 on NK cells and CD8(+) T cells using TC-1 tumor cells transduced with a retrovirus encoding IL-15 linked to IL-15RA (IL-15/IL-15RA). We demonstrated that the expression of IL-15/IL-15RA on TC-1 cells led to increased percentages of tumor-infiltrating NK cells, NKT cells, and CD8(+) T cells, resulting in the inhibition of tumor growth in challenged mice. Additionally, in vivo Ab depletion experiments demonstrated that NK1.1(+) cells and CD8(+) T cells were important in this inhibition of tumor growth. Furthermore, this accumulation of immune cells and inhibition of tumor growth was abolished by a single amino acid mutation in the common gamma-chain binding site on IL-15. We also observed that IL-15/IL-15RA-transduced TC-1 cells led to the activation of STAT5 in NK and CD8(+) T cells in trans, which was abolished in the mutated IL-15/IL-15RA-transduced TC-1 cells. Taken together, our data suggest that common gamma-chain binding-dependent activation of the shared IL-15/IL-2Rbeta/common gamma signaling pathway may play an important role in the activation of NK cells and CD8(+) T cells, resulting in IL-15/IL-15RA trans-presentation-mediated inhibition of tumor growth.


Assuntos
Antígenos Ly/biossíntese , Linfócitos T CD8-Positivos/imunologia , Inibidores do Crescimento/fisiologia , Subunidade gama Comum de Receptores de Interleucina/fisiologia , Interleucina-15/fisiologia , Subunidade beta de Receptor de Interleucina-2/fisiologia , Células Matadoras Naturais/imunologia , Subfamília B de Receptores Semelhantes a Lectina de Células NK/biossíntese , Neoplasias Ovarianas/prevenção & controle , Transdução de Sinais/imunologia , Animais , Apresentação de Antígeno/genética , Antígenos Ly/fisiologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Feminino , Inibidores do Crescimento/biossíntese , Subunidade gama Comum de Receptores de Interleucina/genética , Interleucina-15/biossíntese , Interleucina-15/genética , Interleucina-15/metabolismo , Subunidade alfa de Receptor de Interleucina-15/biossíntese , Subunidade alfa de Receptor de Interleucina-15/genética , Subunidade alfa de Receptor de Interleucina-15/metabolismo , Subunidade alfa de Receptor de Interleucina-15/fisiologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subfamília B de Receptores Semelhantes a Lectina de Células NK/fisiologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/virologia , Retroviridae/genética , Transdução de Sinais/genética , Transdução Genética
10.
Cancer Immunol Immunother ; 58(5): 737-48, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18815785

RESUMO

Current therapeutic approaches to treatment of patients with bulky cervical cancer are based on conventional in situ ablative modalities including cisplatin-based chemotherapy and radiation therapy. The 5-year survival of patients with nonresectable disease is dismal. Because over 99% of squamous cervical cancer is caused by persistent infection with an oncogenic strain of human papillomavirus (HPV), particularly type 16 and viral oncoproteins E6 and E7 are functionally required for disease initiation and persistence, HPV-targeted immune strategies present a compelling opportunity in which to demonstrate proof of principle. Sublethal doses of radiation and chemotherapeutic agents have been shown to have synergistic effect in combination with either vaccination against cancer-specific antigens, or with passive transfer of tumor-specific cytotoxic T lymphocytes (CTLs). Here, we explored the combination of low-dose radiation therapy with DNA vaccination with calreticulin (CRT) linked to the mutated form of HPV-16 E7 antigen (E7(detox)), CRT/E7(detox) in the treatment of E7-expressing TC-1 tumors. We observed that TC-1 tumor-bearing mice treated with radiotherapy combined with CRT/E7(detox) DNA vaccination generated significant therapeutic antitumor effects and the highest frequency of E7-specific CD8(+) T cells in the tumors and spleens of treated mice. Furthermore, treatment with radiotherapy was shown to render the TC-1 tumor cells more susceptible to lysis by E7-specific CTLs. In addition, we observed that treatment with radiotherapy during the second DNA vaccination generated the highest frequency of E7-specific CD8(+) T cells in the tumors and spleens of TC-1 tumor-bearing mice. Finally, TC-1 tumor-bearing mice treated with the chemotherapy in combination with radiation and CRT/E7(detox) DNA vaccination generate significantly enhanced therapeutic antitumor effects. The clinical implications of the study are discussed.


Assuntos
Imunoterapia Ativa , Neoplasias Experimentais/terapia , Vacinas contra Papillomavirus/uso terapêutico , Radioterapia Adjuvante/métodos , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/uso terapêutico , Animais , Apoptose , Calreticulina/genética , Calreticulina/imunologia , Linhagem Celular Transformada/imunologia , Linhagem Celular Transformada/transplante , Transformação Celular Viral , Terapia Combinada , Citocinas/análise , Modelos Animais de Doenças , Feminino , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/radioterapia , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus , Dosagem Radioterapêutica , Proteínas Recombinantes de Fusão/imunologia , Neoplasias do Colo do Útero
11.
J Biomed Sci ; 16: 49, 2009 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-19473507

RESUMO

BACKGROUND: It is important to develop innovative therapies for advanced stage cancers in addition to the conventional therapies including chemotherapy, radiation and surgery. Antigen-specific immunotherapy has emerged as a novel alternate therapy for advanced stage cancers, which may be employed in conjunction with conventional therapies. METHODS: In the current study, we tested the effect of treatment with the chemotherapeutic agent, apigenin in combination with DNA vaccines encoding the HPV-16 E7 antigen linked to heat shock protein 70 (HSP70) in the control of the E7-expressing tumor, TC-1. RESULTS: We observed that treatment with apigenin rendered the TC-1 tumor cells more susceptible to lysis by E7-specific cytotoxic CD8+ T cells. Furthermore, treatment of TC-1 tumor cells with apigenin was found to enhance apoptotic tumor cell death in vitro in a dose-dependant manner. We showed that TC-1 tumor-bearing mice treated with apigenin combined with E7-HSP70 DNA generate highest frequency of primary and memory E7-specific CD8+ T cells, leading to potent therapeutic anti-tumor effects against E7-expressing tumors. CONCLUSION: Thus, apigenin represents a promising chemotherapeutic agent, which may be used in combination with immunotherapy for the treatment of advanced stage cancers. The clinical implications of the current strategy are discussed.


Assuntos
Antineoplásicos/administração & dosagem , Apigenina/uso terapêutico , Imunoterapia/métodos , Animais , Antígenos de Neoplasias/metabolismo , Apoptose , Terapia Combinada/métodos , Feminino , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus , Vacinas contra Papillomavirus/metabolismo , Vacinas de DNA/uso terapêutico
12.
Clin Cancer Res ; 14(10): 3185-92, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18483387

RESUMO

PURPOSE: Because the combination of multiple modalities for cancer treatment is more likely to generate more potent therapeutic effects for the control of cancer, we have explored the combination of chemotherapy using cisplatin, which is routinely used in chemotherapy for advanced cervical cancer, with immunotherapy using DNA vaccines encoding calreticulin (CRT) linked to human papillomavirus type 16 E7 antigen (CRT/E7) in a preclinical model. EXPERIMENTAL DESIGN: We characterized the combination of cisplatin with CRT/E7 DNA vaccine using different regimen for its potential ability to generate E7-specific CD8+ T-cell immune responses as well as antitumor effects against E7-expressing tumors. RESULTS: Our results indicate that treatment of tumor-bearing mice with chemoimmunotherapy combining cisplatin followed by CRT/E7 DNA generated the highest E7-specific CD8+ T-cell immune response and produced the greatest antitumor effects and long-term survival as well as significant levels of E7-specific tumor-infiltrating lymphocytes compared with all the other treatment regimens. Furthermore, we found that treatment with cisplatin leads to the cell-mediated lysis of E7-expressing tumor cells in vitro and increased number of E7-specific CD8+ T-cell precursors in tumor-bearing mice. In addition, we observed that E7-specific CD8+ T cells migrate to and proliferate in the location of TC-1 tumors in mice treated with cisplatin. CONCLUSIONS: Thus, our data suggest that chemoimmunotherapy using cisplatin followed by CRT/E7 DNA vaccine is an effective treatment against E7-expressing tumors and may potentially be translated into the clinical arena.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Cisplatino/uso terapêutico , Neoplasias Experimentais/terapia , Proteínas Oncogênicas Virais/imunologia , Vacinas de DNA/uso terapêutico , Animais , Biolística , Calreticulina/genética , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Terapia Combinada , Citotoxicidade Imunológica , Feminino , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/biossíntese , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Neoplasias Experimentais/imunologia , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus , Reação em Cadeia da Polimerase , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
13.
J Mol Med (Berl) ; 86(8): 899-908, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18542898

RESUMO

There is an urgent need to develop new innovative therapies for the control of cancer. Antigen-specific immunotherapy and the employment of proteasome inhibitors have emerged as two potentially plausible approaches for the control of cancer. In the current study, we explored the combination of the DNA vaccine encoding calreticulin (CRT) linked to human papillomavirus type 16 E7 antigen (CRT/E7) with the proteasome inhibitor, bortezomib, for their ability to generate E7-specific immune responses and antitumor effects in vaccinated mice. We found that the combination of treatment with bortezomib and CRT/E7(detox) DNA generated more potent E7-specific CD8+ T cell immune responses and better therapeutic effects against TC-1 tumors in tumor-bearing mice compared to monotherapy. Furthermore, we found that treatment with bortezomib led to increased apoptosis of TC-1 tumor cells and could render the TC-1 tumor cells more susceptible to lysis by E7-specific CD8+ T cells. Our data have significant implications for future clinical translation.


Assuntos
Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Inibidores de Proteases/uso terapêutico , Inibidores de Proteassoma , Pirazinas/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/uso terapêutico , Animais , Apoptose , Bortezomib , Calreticulina/genética , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Quimioterapia Combinada , Camundongos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Células Tumorais Cultivadas , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
14.
J Formos Med Assoc ; 107(3): 198-217, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18400605

RESUMO

Human papillomavirus (HPV) has been considered to be an etiologic factor for anogenital cancers, such as cervical cancer and possibly a subset of cancers of the aerodigestive tract. These small, non-enveloped, double-stranded DNA viruses primarily infect the epithelium and induce benign as well as malignant lesions of the mucosa and skin. Some HPVs are considered to be high-risk due to their strong implication in carcinogenesis, particularly the malignant progression of cervical tumors. The recognition of papillomaviruses as a major etiologic agent for human cancers has increased their medical importance and stimulated research into developing strategies for the screening, diagnosis, prevention and treatment of HPV-associated diseases.


Assuntos
Papillomaviridae/genética , Infecções por Papillomavirus/epidemiologia , Infecções Tumorais por Vírus/epidemiologia , Neoplasias do Ânus/etiologia , Condiloma Acuminado/etiologia , Feminino , Doenças dos Genitais Femininos/virologia , Humanos , Neoplasias Laríngeas/etiologia , Masculino , Papillomaviridae/classificação , Neoplasias Cutâneas/etiologia , Neoplasias do Colo do Útero/etiologia
15.
Curr Mol Med ; 7(5): 490-503, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17691964

RESUMO

Human papillomavirus (HPV) is a causal agent for approximately 5.3% of cancers worldwide, including cervical cancer, and subsets of genital and head and neck cancer. Persistent HPV infection is a necessary, but not sufficient, cause of cervical cancer. Of the >100 HPV genotypes, only about a dozen, termed "high-risk", are associated with cancer. HPV-16 is present in approximately 50% of all cervical cancers and HPV-16, HPV-18, HPV-31 and HPV-45 together account for approximately 80%. Most high-risk HPV infections are subclinical, and are cleared by the host's immune system. The remainder produces low or high-grade squamous intraepithelial lesions (SILs), also called cervical intraepithelial neoplasia (CIN), which also may regress spontaneously. However persistent high grade SIL represents the precursor lesion of cervical cancer and carcinogenic progression is associated with integration of the viral DNA, loss of E2 and upregulation of viral oncogene expression, and chromosomal rearrangements like 3q gain. Cytologic screening of the cervix for SIL and intervention has reduced the incidence of cervical cancer in the US by an estimated 80% and HPV viral DNA and other molecular tests may improve screening further. The licensure of a preventive HPV vaccine ushers in a new era, but issues remain, including: protection restricted to a few oncogenic HPV types, access in low resource settings and impact on current cytologic screening protocols. Importantly, preventive HPV vaccination does not help with current HPV infection or disease. Here we examine the potential of second-generation preventive HPV vaccines and therapeutic HPV vaccination to address these outstanding issues.


Assuntos
Neoplasias do Colo do Útero/prevenção & controle , Neoplasias do Colo do Útero/terapia , Alphapapillomavirus/genética , Alphapapillomavirus/fisiologia , Animais , Vacinas Anticâncer/imunologia , Feminino , Genes Virais , Humanos , Programas de Rastreamento , Replicação Viral
16.
Immunol Lett ; 114(2): 86-93, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17976741

RESUMO

DNA vaccination has emerged as a promising strategy for cancer immunotherapy. However, since DNA vaccines have low immunogenicity, various strategies have been developed to enhance the potency of DNA vaccines. In the current study, we aim to determine whether the potency of the DNA vaccine encoding human papillomavirus type 16 (HPV-16) E7 antigen can be enhanced by IL-2. We have generated a DNA vaccine encoding IL-2 linked to HPV-16 E7 antigen. Our results indicate that the DNA vaccine encoding a fusion of IL-2 and E7 proteins generated the highest frequency of E7-specific CD8(+) T cells. We also found that the DNA vaccine encoding a fusion of IL-2 and E7 proteins generated the strongest protective as well as therapeutic anti-tumor effect against E7-expressing tumors. In addition, it was observed that CD8(+) T cells were mainly responsible for the antitumor effect generated by the DNA vaccine encoding a fusion of IL-2 and E7 proteins. Thus, we conclude that the linkage of IL-2 to HPV-16 E7 antigen significantly enhances the DNA vaccine potency against E7-expressing tumors. Our strategy may potentially be used in other antigenic systems to control infectious diseases and/or cancer.


Assuntos
Vacinas Anticâncer/imunologia , Papillomavirus Humano 16/imunologia , Interleucina-2/imunologia , Neoplasias Experimentais/prevenção & controle , Proteínas E7 de Papillomavirus/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia , Animais , Vacinas Anticâncer/genética , Interleucina-2/genética , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/virologia , Proteínas E7 de Papillomavirus/genética , Vacinas contra Papillomavirus/genética , Vacinas contra Papillomavirus/imunologia , Proteínas Recombinantes de Fusão , Linfócitos T Citotóxicos/metabolismo , Vacinas de DNA/genética
17.
Curr Opin Investig Drugs ; 8(12): 1038-50, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18058574

RESUMO

Cervical cancer is the second leading cause of cancer deaths in women worldwide and HPV infection is responsible for the development of this cancer. Effective vaccination against HPV represents an opportunity for the control of cervical cancer. The newly licensed preventive HPV vaccine in the US, Gardasil, has both a good safety profile and clinical efficacy against the HPV genotypes from which it was derived. However, this vaccine can only protect against up to 70 to 80% of cervical cancer and also lacks therapeutic efficacy against established HPV infection and HPV-associated lesions. Thus, the future of HPV vaccination needs to focus on the development of a new generation of preventive and therapeutic vaccines that are capable of protecting against most cervical cancers.


Assuntos
Infecções por Papillomavirus/prevenção & controle , Vacinas contra Papillomavirus , Neoplasias do Colo do Útero/prevenção & controle , Neoplasias do Colo do Útero/virologia , Vacinas Virais/uso terapêutico , Ensaios Clínicos como Assunto , Feminino , Humanos , Modelos Imunológicos , Vacinas contra Papillomavirus/administração & dosagem , Vacinas contra Papillomavirus/efeitos adversos , Vacinas contra Papillomavirus/imunologia
18.
Exp Mol Med ; 39(6): 679-89, 2007 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-18160838

RESUMO

More than 99% of cervical cancers have been associated with human papillomaviruses (HPVs), particularly HPV type 16. The clear association between HPV infection and cervical cancer indicates that HPV serves as an ideal target for development of preventive and therapeutic vaccines. Although the recently licensed preventive HPV vaccine, Gardasil, has been shown to be safe and capable of generating significant protection against specific HPV types, it does not have therapeutic effect against established HPV infections and HPV-associated lesions. Two HPV oncogenic proteins, E6 and E7, are consistently co-expressed in HPV-expressing cervical cancers and are important in the induction and maintenance of cellular transformation. Therefore, immunotherapy targeting E6 and/or E7 proteins may provide an opportunity to prevent and treat HPV-associated cervical malignancies. It has been established that T cell-mediated immunity is one of the most crucial components to defend against HPV infections and HPV-associated lesions. Therefore, effective therapeutic HPV vaccines should generate strong E6/E7-specific T cell-mediated immune responses. DNA vaccines have emerged as an attractive approach for antigen-specific T cell-mediated immunotherapy to combat cancers. Intradermal administration of DNA vaccines via a gene gun represents an efficient way to deliver DNA vaccines into professional antigen-presenting cells in vivo. Professional antigen-presenting cells, such as dendritic cells, are the most effective cells for priming antigen-specific T cells. Using the gene gun delivery system, we tested several DNA vaccines that employ intracellular targeting strategies for enhancing MHC class I and class II presentation of encoded model antigen HPV-16 E7. Furthermore, we have developed a strategy to prolong the life of DCs to enhance DNA vaccine potency. More recently, we have developed a strategy to generate antigen-specific CD4(+) T cell immune responses to further enhance DNA vaccine potency. The impressive pre- clinical data generated from our studies have led to several HPV DNA vaccine clinical trials.


Assuntos
Papillomaviridae/genética , Infecções por Papillomavirus/prevenção & controle , Vacinas contra Papillomavirus/administração & dosagem , Neoplasias do Colo do Útero/prevenção & controle , Vacinas de DNA/administração & dosagem , Feminino , Humanos , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/imunologia , Papillomaviridae/imunologia , Infecções por Papillomavirus/imunologia , Proteínas Repressoras , Vacinas Virais/administração & dosagem
19.
Vaccine ; 29(5): 1082-9, 2011 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-21130752

RESUMO

Protein-based vaccines have emerged as a potentially promising approach for the generation of antigen-specific immune responses. However, due to their low immunogenicity, there is a need for innovative approaches to enhance protein-based vaccine potency. One approach to enhance protein-based vaccine potency is the employment of toll-like receptor ligands, such as CpG oligonucleotides, to activate the antigen-specific T cell immune responses. Another approach involves employing a method capable of improving the delivery of protein-based vaccine intramuscularly to lead to the slow release of the protein, resulting in improved vaccine potency. In the current study, we aimed to determine whether intramuscular injection of protein-based vaccines in conjunction with CpG followed by electroporation can lead to increased delivery of the protein-based vaccine into muscle cells, resulting in enhanced protein-based vaccine potency. We found that intramuscular injection followed by electroporation can effectively transduce the protein-based vaccine into the muscle cells. Furthermore, we found that intramuscular vaccination with OVA protein in combination with CpG followed by electroporation generates the best OVA-specific CD8+ T cell immune responses as well as the best protective and therapeutic antitumor effects in vaccinated mice. CD8+ T cells were found to play an important role in the observed protective antitumor effects generated by the vaccination. Similar results were observed using the HPV-16 E7 protein-based vaccination system. Thus, our data indicate that intramuscular administration of protein-based vaccines in conjunction with CpG followed by electroporation can significantly enhance the antigen-specific CD8+ T cell immune responses. The clinical implications of the study are discussed.


Assuntos
Eletroporação , Injeções Intramusculares , Ovalbumina/imunologia , Proteínas E7 de Papillomavirus/imunologia , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Ovalbumina/administração & dosagem , Proteínas E7 de Papillomavirus/administração & dosagem , Infecções por Papillomavirus/prevenção & controle , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
20.
Hum Gene Ther ; 22(7): 809-19, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21128743

RESUMO

Abstract Antigen-specific immunotherapy and vascular disrupting agents, such as 5,6-dimethylxanthenone-4-acetic acid (DMXAA), have emerged as attractive approaches for the treatment of cancers. In the current study, we tested the combination of DMXAA treatment with therapeutic human papillomavirus type 16 (HPV-16) E7 peptide-based vaccination for their ability to generate E7-specific CD8+ T-cell immune responses, as well as their ability to control E7-expressing tumors in a subcutaneous and a cervicovaginal tumor model. We found that the combination of DMXAA treatment with E7 long peptide (amino acids 43-62) vaccination mixed with polyriboinosinic:polyribocytidylic generated significantly stronger E7-specific CD8+ T-cell immune responses and antitumor effects compared with treatment with DMXAA alone or HPV peptide vaccination alone in the subcutaneous model. Additionally, we found that the DMXAA-mediated enhancement of E7-specific CD8+ T-cell immune responses generated by the therapeutic HPV peptide-based vaccine was dependent on the timing of administration of DMXAA. Treatment with DMXAA in tumor-bearing mice was also shown to lead to increased dendritic cell maturation and increased production of inflammatory cytokines in the tumor. Furthermore, we observed that the combination of DMXAA with HPV-16 E7 peptide vaccination generated a significant enhancement in the antitumor effects in the cervicovaginal TC-1 tumor growth model, which closely resembles the tumor microenvironment of cervical cancer. Taken together, our data demonstrated that administration of the vascular disrupting agent, DMXAA, enhances therapeutic HPV vaccine-induced cytotoxic T-lymphocyte responses and antitumor effects against E7-expressing tumors in two different locations. Our study has significant implications for future clinical translation.


Assuntos
Proteínas E7 de Papillomavirus/genética , Vacinas contra Papillomavirus/administração & dosagem , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus/imunologia , Vacinas contra Papillomavirus/imunologia , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/virologia , Xantonas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa