Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 29(20): 3646-54, 2011 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-21435405

RESUMO

HER2/neu is an oncogene amplified and over-expressed in 20-30% of breast adenocarcinomas. Treatment with the humanized monoclonal antibody trastuzumab has shown efficacy in combination with cytotoxic agents, although resistance occurs over time. Novel approaches are needed to further increase antibody efficacy. In this study, we provide evidence in a mouse breast cancer therapeutic tumor model that the combination of active immunization with a modified HER2/neu DNA vaccine and passive infusion of an anti-HER2/neu monoclonal antibody leads to significant regression of established tumors. Our data indicate that combination therapy with a HER2/neu DNA vaccine and trastuzumab may have clinical activity in breast cancer patients.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Vacinas Anticâncer/imunologia , Epitopos/imunologia , Neoplasias Mamárias Experimentais/terapia , Receptor ErbB-2/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/imunologia , Vacinas Anticâncer/administração & dosagem , Feminino , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Trastuzumab , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
2.
Mod Pathol ; 21(6): 727-32, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18344976

RESUMO

Prostate-specific membrane antigen is a type II membrane glycoprotein that is expressed in benign and neoplastic prostatic tissue and has been recently shown to be also expressed in the neovasculature of various solid malignant tumors including renal cell carcinoma. Renal cell carcinoma is a heterogeneous group of tumors with distinct morphologic and genetic characteristics and clinical behaviors. We performed immunohistochemical studies on formalin-fixed, paraffin-embedded archival material from 75 nephrectomies, using antibodies 13D6 against prostate-specific membrane antigen and CD31 against endothelial cells. The study included 30 clear cell renal cell carcinomas, and 15 of each of papillary and chromophobe renal cell carcinoma and oncocytoma. The extent and intensity of staining were assessed semiquantitatively. In all cases, immunoreactivity was detected only in the tumor-associated neovasculature and not in tumor cells. Clear cell renal cell carcinoma showed the most diffuse staining pattern, where 24/30 cases or 80% had >50% reactive vessels, followed by chromophobe renal cell carcinoma (9/15; 60%) and oncocytoma (5/15, 33%). No diffuse staining was detected in any of the papillary renal cell carcinomas and only focal staining was detected in 11 cases (11/15; 73%). Staining intensity was the strongest in clear cell renal cell carcinoma (25/30; 83%) followed by chromophobe renal cell carcinoma (9/15; 60%), oncocytoma (8/15, 53%) and papillary renal cell carcinoma (5/15; 33%). In summary, prostate-specific membrane antigen is expressed in tumor-associated neovasculature of the majority of renal cortical tumors and is most diffusely and intensely expressed in clear cell renal cell carcinoma and least in papillary renal cell carcinoma. The differences in the expression of prostate-specific membrane antigen in renal cell carcinoma subtypes provide further evidence of the biological diversity of these tumors, and diagnostic and therapeutic applications of such expression can be expanded to include subtypes of renal cell carcinoma.


Assuntos
Antígenos de Superfície/biossíntese , Biomarcadores Tumorais/análise , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Glutamato Carboxipeptidase II/biossíntese , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Carcinoma de Células Renais/irrigação sanguínea , Células Endoteliais/metabolismo , Humanos , Imuno-Histoquímica , Neoplasias Renais/irrigação sanguínea
3.
Clin Cancer Res ; 13(20): 6195-203, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17947487

RESUMO

PURPOSE: HER2/neu is frequently overexpressed in breast cancer. In a mouse model, vaccination with HER2/neu DNA elicits antibodies that confer partial protection against tumor challenge. EXPERIMENTAL DESIGN: To enhance antitumor immunity, we fused cDNA encoding Flt-3 ligand (FL) to the rat HER2/neu extracellular domain (neu), generating a chimeric FLneu molecule. FLneu and neu DNA vaccines were compared for immunogenicity and their ability to protect mice from tumor challenge. RESULTS: The neu vaccine generated a HER2/neu-specific antibody response. In contrast, vaccination with FLneu induced CD8+ T cells specific for HER2/neu but a negligible anti-HER2/neu antibody response. The switch from an antibody-mediated to T cell-mediated response was due to different intracellular localization of neu and FLneu. Although the neu protein was secreted, the FLneu protein was retained inside the cell, co-localizing with the endoplasmic reticulum, facilitating processing and presentation to T cells. The neu and FLneu vaccines individually conferred only weak tumor immunity. However, efficient tumor rejection was seen when neu and FLneu were combined, inducing both strong anti-HER2/neu-specific antibody and T cell responses. Adoptive transfer of both immune CD8+ T cells and immune sera from immunized mice was required to confer tumor immunity in naïve hosts. CONCLUSIONS: These results show that active induction of both humoral and cellular immunity to HER2/neu is required for efficient tumor protection, and that neither response alone is sufficient.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer , DNA/química , Proteínas de Membrana/química , Neuraminidase/química , Animais , Linfócitos T CD8-Positivos/imunologia , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Humanos , Ligantes , Camundongos , Microscopia de Fluorescência , Transplante de Neoplasias , Neuraminidase/metabolismo , Estrutura Terciária de Proteína , Linfócitos T/metabolismo , Fatores de Tempo , Vacinas de DNA/química
4.
Clin Cancer Res ; 12(18): 5511-9, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17000687

RESUMO

PURPOSE: Plasmid DNAs encoding cytokines enhance immune responses to vaccination in models of infectious diseases and cancer. We compared DNA adjuvants for their ability to enhance immunity against a poorly immunogenic self-antigen expressed by cancer. EXPERIMENTAL DESIGN: DNAs encoding cytokines that affect T cells [interleukin (IL)-2, IL-12, IL-15, IL-18, IL-21, and the chemokine CCL21] and antigen-presenting cells [granulocyte macrophage colony-stimulating factor (GM-CSF)] were compared in mouse models as adjuvants to enhance CD8+ T-cell responses and tumor immunity. A DNA vaccine against a self-antigen, gp100, expressed by melanoma was used in combination with DNA encoding cytokines and cytokines fused to the Fc domain of mouse IgG1 (Ig). RESULTS: We found that (a) cytokine DNAs generally increased CD8+ T-cell responses against gp100; (b) ligation to Fc domains further enhanced T-cell responses; (c) adjuvant effects were sensitive to timing of DNA injection; (d) the most efficacious individual adjuvants for improving tumor-free survival were IL-12/Ig, IL-15/Ig, IL-21/Ig, GM-CSF/Ig, and CCL21; and (e) combinations of IL-2/Ig+IL-12/Ig, IL-2/Ig+IL-15/Ig, IL-12/Ig+IL-15/Ig, and IL-12/Ig+IL-21/Ig were most active; and (f) increased adjuvanticity of cytokine/Ig fusion DNAs was not related to higher tissue levels or greater stability. CONCLUSIONS: These observations support the potential of cytokine DNA adjuvants for immunization against self-antigens expressed by cancer, the importance of timing, and the enhancement of immune responses by Fc domains through mechanisms unrelated to increased half-life.


Assuntos
Adjuvantes Imunológicos/farmacologia , Citocinas/genética , Imunoterapia Ativa/métodos , Neoplasias/imunologia , Vacinas de DNA/uso terapêutico , Reação de Fase Aguda/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Animais , Autoantígenos/imunologia , Antígenos CD8/metabolismo , Citocinas/imunologia , Rejeição de Enxerto/imunologia , Hipopigmentação/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Imunoterapia Ativa/efeitos adversos , Imunoterapia Ativa/estatística & dados numéricos , Interleucina-12/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/química , Plasmídeos/imunologia , Análise de Sobrevida , Linfócitos T/imunologia , Fatores de Tempo , Vacinas de DNA/efeitos adversos , Vacinas de DNA/química , Vacinas de DNA/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
5.
Int J Cancer ; 116(3): 415-21, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15800947

RESUMO

Prostate-specific membrane antigen (PSMA) is a prototypical differentiation antigen expressed on normal and neoplastic prostate epithelial cells, and on the neovasculature of many solid tumors. Monoclonal antibodies specific for PSMA are in development as therapeutic agents. Methodologies to actively immunize against PSMA may be limited by immunologic ignorance and/or tolerance that restrict the response to self-antigens. Our studies have previously shown that xenogeneic immunization with DNA vaccines encoding melanosomal differentiation antigens induces immunity in a mouse melanoma model. Here we apply this approach to PSMA to establish proof of principle in a mouse model. Immunization with xenogeneic human PSMA protein or DNA induced antibodies to both human and mouse PSMA in mice. Monoclonal antibodies specific for mouse PSMA were generated to analyze antibody isotypes and specificity for native and denatured PSMA at the clonal level. Most antibodies recognized denatured PSMA, but C57BL/6 mice immunized with xenogeneic PSMA DNA followed by a final boost with xenogeneic PSMA protein yielded autoantibodies that reacted with native folded mouse PSMA. Monoclonal antibodies were used to confirm the expression of PSMA protein in normal mouse kidney. These results establish the basis for clinical trials to test PSMA DNA vaccines in patients with solid tumors that either express PSMA directly or that depend on normal endothelial cells expressing PSMA for their continued growth.


Assuntos
Anticorpos Heterófilos/imunologia , Antígenos de Superfície/biossíntese , Antígenos de Superfície/imunologia , Autoanticorpos/imunologia , Glutamato Carboxipeptidase II/biossíntese , Glutamato Carboxipeptidase II/imunologia , Neoplasias da Próstata/imunologia , Animais , Formação de Anticorpos , Modelos Animais de Doenças , Células Endoteliais , Perfilação da Expressão Gênica , Humanos , Imunização/veterinária , Imunoterapia/métodos , Masculino , Camundongos , Neoplasias da Próstata/veterinária , Vacinas de DNA
6.
Vaccine ; 22(13-14): 1700-8, 2004 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-15068853

RESUMO

Xenogeneic DNA vaccination can elicit tumor immunity through T cell and antibody-dependent effector mechanisms. Blockade of CTLA-4 engagement with B7 expressed on APCs has been shown to enhance T cell-dependent immunity. We investigated whether CTLA-4 blockade could increase T-cell responses and tumor immunity elicited by DNA vaccines. CTLA-4 blockade enhanced B16 tumor rejection in mice immunized against the melanoma differentiation antigens tyrosinase-related protein 2 and gp100, and this effect was stronger when anti-CTLA-4 was administered with booster vaccinations. CTLA-4 blockade also increased the T-cell responses to prostate-specific membrane antigen (PSMA) when given with the second or third vaccination. Based on these pre-clinical studies, we suggest that anti-CTLA-4 should be tested with xenogeneic DNA vaccines against cancer and that special attention should be given to sequence and schedule of administration.


Assuntos
Antígenos de Diferenciação/farmacologia , Autoantígenos/imunologia , Autoimunidade/imunologia , Vacinas Anticâncer/imunologia , Imunidade Celular/imunologia , Imunossupressores/farmacologia , Neoplasias/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Antineoplásicos/biossíntese , Antígenos CD , Antígenos de Diferenciação/imunologia , Antígeno CTLA-4 , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Esquemas de Imunização , Masculino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Plasmídeos/imunologia , Próstata/imunologia , Vacinas de DNA/imunologia
7.
Semin Oncol ; 30(5): 659-66, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14571413

RESUMO

Immunotherapy is currently being investigated as a treatment for patients with asymptomatic, recurrent prostate cancer manifested only by a rising prostate-specific antigen (PSA) level. Several different approaches to active immunization against antigens found on cancer cells have been explored. Immunization with DNA overcomes many of the obstacles noted in previous studies. Injection of plasmid DNA encoding a xenogeneic differentiation antigen (prostate-specific membrane antigen [PSMA]) is a potent means to induce antibody and T-cell responses to these otherwise poorly immunogenic self proteins. Use of the xenogeneic DNA (ie, human PSMA DNA injected into mouse) has been shown to be an absolute requirement to overcome immunologic tolerance. We are currently conducting a phase I trial of human and mouse PSMA DNA vaccines in patients with recurrent prostate cancer, based on preclinical experiments described below.


Assuntos
Recidiva Local de Neoplasia/terapia , Neoplasias da Próstata/terapia , Vacinação/métodos , Vacinas de DNA/uso terapêutico , Animais , Antígenos Heterófilos/imunologia , Antígenos Heterófilos/uso terapêutico , Antígenos de Superfície/imunologia , Antígenos de Superfície/uso terapêutico , Antígenos Glicosídicos Associados a Tumores/imunologia , Antígenos Glicosídicos Associados a Tumores/uso terapêutico , Autoantígenos/imunologia , Biomarcadores Tumorais/sangue , Ensaios Clínicos Fase I como Assunto , Células Dendríticas/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Glutamato Carboxipeptidase II/imunologia , Glutamato Carboxipeptidase II/uso terapêutico , Proteínas de Choque Térmico/imunologia , Humanos , Tolerância Imunológica/imunologia , Masculino , Camundongos , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/imunologia , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/imunologia , Linfócitos T/imunologia , Vacinas de DNA/classificação , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Vacinas Virais/uso terapêutico
8.
Semin Cancer Biol ; 12(1): 63-71, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11926414

RESUMO

Cancer poses a difficult problem for immunotherapy because it arises from the host's own tissues. Many of the target antigens are tissue-specific molecules shared by cancer cells and normal cells. Thus, these are weak antigens that do not typically elicit immunity. In addition, tumors have several features that make their recognition and destruction by the immune system difficult. Despite these obstacles, several strategies for developing effective tumor immunity have been developed. Crucial to these approaches is the discovery and understanding of the molecular identity of antigens and the mechanisms involved in tumor immunity. In this review, strategies to overcome immune ignorance and tolerance are discussed.


Assuntos
Tolerância Imunológica , Neoplasias/imunologia , Antígenos de Neoplasias/imunologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...