Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Eur J Immunol ; 44(8): 2331-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24810893

RESUMO

NK cells that mediate ADCC play an important role in tumor-specific immunity. We have examined factors limiting specific lysis of tumor cells by CD16.NK-92 cells induced by CNTO 95LF antibodies recognizing αV integrins that are overexpressed on many tumor cells. Although all tested tumor cells were killed by CD16.NK-92 effectors in the presence of the antibodies, the killing of target cells with a low level of ICAM-1 expression revealed a dramatic decrease in their specific lysis at high antibody concentration, revealing a dose limiting effect. A similar effect was also observed with primary human NK cells. The effect was erased after IFN-γ treatment of tumor cells resulting in upregulation of ICAM-1. Furthermore, killing of the same tumor cells induced by Herceptin antibody was significantly impaired in the presence of CNTO 95Ala-Ala antibody variant that blocks αV integrins but is incapable of binding to CD16. These data suggest that αV integrins on tumor cells could compensate for the loss of ICAM-1 molecules, thereby facilitating ADCC by NK cells. Thus, NK cells could exercise cytolytic activity against ICAM-1 deficient tumor cells in the absence of proinflammatory cytokines, emphasizing the importance of NK cells in tumor-specific immunity at early stages of cancer.


Assuntos
Anticorpos/imunologia , Integrina alfaV/imunologia , Células Matadoras Naturais/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Citocinas/imunologia , Citotoxicidade Imunológica , Proteínas Ligadas por GPI/imunologia , Humanos , Inflamação/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Interferon gama/imunologia , Receptores de IgG/imunologia , Células Tumorais Cultivadas , Regulação para Cima/imunologia
2.
Genes Chromosomes Cancer ; 53(4): 349-57, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24488787

RESUMO

Intetumumab is a fully human monoclonal antibody that inhibits αv integrins. It has been shown in in vitro assays to effectively inhibit cell viability, metastasis, and adhesion of human cancer cells and endothelial cells. However, the response to Intetumumab varies in different tumor cell lines. To understand the growth inhibition mechanism of Intetumumab and to identify a molecular signature that can predict sensitivity, we focused on lung cancer cell lines and performed a series of proliferation assays. We then assessed the global gene expression profiles, DNA copy number variations, and microRNA profiles from a total of 23 lung cancer lines. The results revealed that lung cancer sensitivity to Intetumumab is associated with several chromosomal alterations, particularly genetic loss at chromosome arm 19p, which resulted in gene expression changes. We identified a genetic signature that can be used to predict Intetumumab sensitivity for lung cancer cell lines. Independently, microRNA analysis revealed a panel of signature microRNAs that includes several markers of epithelial to mesenchymal transition and tumor metastasis such as miR-200 family and miR-205. Both the genetic and microRNA signatures provide insights into the potential mechanism of Intetumumab activity and serve as the first step to develop a patient stratification strategy for Intetumumab therapy in lung cancer.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Integrina alfaV/metabolismo , Neoplasias Pulmonares/genética , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral/efeitos dos fármacos , Cromossomos Humanos Par 19/genética , Variações do Número de Cópias de DNA , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , MicroRNAs/genética , Metástase Neoplásica
3.
Anticancer Drugs ; 24(3): 237-50, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23275294

RESUMO

Both Src and αV integrins are important for tumor growth and angiogenesis. They are interconnected and responsible for important features of the tumor phenotype including invasiveness, metastasis, angiogenesis, and resistance to apoptosis. This study examines whether combinational inhibition of both integrin and Src pathways would exert greater antiangiogenesis and antitumor effects than either pathway alone. Using in-vitro cell culture systems, the activity of CNTO95 (Intetumumab), an αV integrin inhibitor, and dasatinib, an Src inhibitor, on proliferation, adhesion, and migration was evaluated in colon cancer cell lines, HCT-116 and RKO, as well as HUVEC cells. The antiangiogenic effect of this combinatory regimen was also tested using an in-vitro tubular network formation assay. The effects of CNTO95 and dasatinib on the activation of Src and integrin pathway signal transduction were also determined by western blotting. The combination of CNTO95 plus dasatinib inhibited adhesion, migration, and paxillin phosphorylation in both HCT-116 and RKO cells. CNTO95 and dasatinib also led to increased apoptosis of HCT-116 cells; however, similar effects were not observed in RKO cells. In addition, dual treatment of CNTO95 and dasatinib exerted enhanced effects on HUVEC cell proliferation, invasion, tubular network formation, and paxillin phosphorylation. In conclusion, our results suggest that concurrent inhibition of both the integrin and the Src pathways exert more pronounced antiangiogenic and antitumor effects than with either pathway being inhibited alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Integrina alfaV/metabolismo , Quinases da Família src/antagonistas & inibidores , Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Dasatinibe , Quinase 1 de Adesão Focal/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Paxilina/metabolismo , Fosforilação/efeitos dos fármacos , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tiazóis/administração & dosagem , Tiazóis/farmacologia , Quinases da Família src/metabolismo
4.
J Neurooncol ; 110(1): 27-36, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22842979

RESUMO

Brain metastases commonly occur in patients with breast, lung and melanoma systemic cancers. The anti-α(V) integrin monoclonal antibody intetumumab binds cell surface proteins important for adhesion, invasion and angiogenesis in the metastatic cascade. The objective of this study was to investigate the anti-metastatic effect of intetumumab in a hematogenous breast cancer brain metastasis model. Female nude rats received intra-carotid infusion of human brain-seeking metastatic breast cancer cells (231BR-HER2) and were randomly assigned into four groups: (1) control; (2) intetumumab mixed with cells in vitro 5 min before infusion without further treatment; (3) intetumumab intravenously 4 h before and weekly after cell infusion; (4) intetumumab intravenously weekly starting 7 days after cell infusion. Brain metastases were detected by magnetic resonance imaging (MRI) and immunohistochemistry. Comparisons were made using the Kruskal-Wallis test and Dunnett's test. Survival times were estimated using Kaplan-Meier analysis. All control rats with brain tissue available for histology (9 of 11 rats) developed multiple brain metastases (median = 14). Intetumumab treatment either in vitro prior to cell infusion or intravenous before or after cell infusion prevented metastasis formation on MRI and decreased the number of metastases on histology (median = 2, p = 0.0055), including 30 % of animals without detectable tumors at the end of the study. The overall survival was improved by intetumumab compared to controls (median 77+ vs. 52 days, p = 0.0277). Our results suggest that breast cancer patients at risk of metastases might benefit from early intetumumab treatment.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Integrina alfa5/metabolismo , Animais , Anticorpos Monoclonais Humanizados , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Invasividade Neoplásica/patologia , Ratos , Ratos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Int J Gynecol Cancer ; 21(6): 1084-90, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21633302

RESUMO

OBJECTIVE: Uterine serous papillary carcinoma (USPC) is an aggressive variant of endometrial cancer characterized by an innate resistance to chemotherapy and poor prognosis. In this study, we evaluated the expression of αV-integrins in primary USPC cell lines and the in vitro ability of intetumumab (CNTO 95), a fully human monoclonal antibody against αV-integrins, to inhibit USPC cell adhesion and migration. MATERIALS AND METHODS: The surface expression of integrins belonging to the αV-family, including αVß3, αVß5, and αVß6, was evaluated in 6 primary USPC cell lines using flow cytometry analysis. To test the ability of intetumumab to inhibit USPC cell adhesion and migration, adhesion assays in the presence of vitronectin and migration assays through an 8.0-µm pore polycarbonate membrane also were performed. RESULTS: We found high expression of the αV-subunit on the cell surface of all 6 primary USPC cell lines tested (100% positive cells; mean fluorescence intensity range, 13.1-39.5). When the expression of single heterodimeric integrins was evaluated, αVß3, αVß5, and αVß6 were expressed on 37.5%, 32.0%, and 16.3% of cells (mean fluorescence intensity range, 6.5-16.2, 9.2-32.5, and 6.2-11.5, respectively). Importantly, in functional assays, low doses of intetumumab were effective in inhibiting adhesion (0.15 µg/mL, P = 0.003) and migration (1.25 µg/mL P = 0.02) of primary USPC cell lines. CONCLUSIONS: The αV-integrins are overexpressed on the cell surface of primary USPC cell lines. Intetumumab may significantly inhibit USPC cell adhesion and migration pathways and may therefore represent a novel treatment option for patients harboring this rare but highly aggressive variant of endometrial cancer.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Integrina alfaV/metabolismo , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/metabolismo , Idoso , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral/metabolismo , Cistadenocarcinoma Papilar/tratamento farmacológico , Cistadenocarcinoma Papilar/metabolismo , Cistadenocarcinoma Papilar/patologia , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Feminino , Citometria de Fluxo , Humanos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Neoplasias Uterinas/patologia
6.
Mol Cancer Ther ; 5(2): 347-55, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16505108

RESUMO

We investigated the significance of erythropoietin receptor (EPOR) expression following treatment with recombinant human erythropoietin (rHuEPO; epoetin alpha) and the effect of recombinant epoetins (epoetin alpha, epoetin beta, and darbepoetin alpha) alone or in combination with anticancer therapy on tumor growth in two well-established preclinical models of breast carcinoma (MDA-MB-231 and MCF-7 cell lines). Expression and localization of EPOR under hypoxic and normoxic conditions in MDA-MB-231 and MCF-7 cells were evaluated by immunoblotting, flow cytometry, and immunohistochemistry. EPOR binding was evaluated using [125I]rHuEPO. Proliferation, migration, and signaling in MDA-MB-231 and MCF-7 cells following treatment with rHuEPO were evaluated. Tumor growth was assessed following administration of recombinant epoetins alone and in combination with paclitaxel (anticancer therapy) in orthotopically implanted MDA-MB-231 and MCF-7 breast carcinoma xenograft models in athymic mice. EPOR expression was detected in both tumor cell lines. EPOR localization was found to be exclusively cytosolic and no specific [125I]rHuEPO binding was observed. There was no stimulated migration, proliferation, or activation of mitogen-activated protein kinase and AKT following rHuEPO treatment. In mice, treatment with recombinant epoetins alone and in combination with paclitaxel resulted in equivalent tumor burdens compared with vehicle-treated controls. Results from our study suggest that although EPOR expression was observed in two well-established breast carcinoma cell lines, it was localized to a cytosolic distribution and did not transduce a signaling cascade in tumors that leads to tumor growth. The addition of recombinant epoetins to paclitaxel did not affect the outcome of paclitaxel therapy in breast carcinoma xenograft models. These results show that recombinant epoetins do not evoke a physiologic response on EPOR-bearing tumor cells as assessed by numerous variables, including growth, migration, and cytotoxic challenge in preclinical in vivo tumor models.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Eritropoetina/uso terapêutico , Receptores da Eritropoetina/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/química , Neoplasias da Mama/metabolismo , Carcinoma/química , Carcinoma/metabolismo , Hipóxia Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Eritropoetina/efeitos adversos , Feminino , Humanos , Camundongos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Paclitaxel/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores da Eritropoetina/análise , Proteínas Recombinantes , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Neuro Oncol ; 13(1): 51-60, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21123368

RESUMO

We used dynamic MRI to evaluate the effects of monoclonal antibodies targeting brain tumor vasculature. Female athymic rats with intracerebral human tumor xenografts were untreated or treated with intetumumab, targeting α(V)-integrins, or bevacizumab, targeting vascular endothelial growth factor (n = 4-6 per group). Prior to treatment and at 1, 3, and 7 days after treatment, we performed standard MRI to assess tumor volume, dynamic susceptibility-contrast MRI with the blood-pool iron oxide nanoparticle ferumoxytol to evaluate relative cerebral blood volume (rCBV), and dynamic contrast-enhanced MRI to assess tumor vascular permeability. Tumor rCBV increased by 27 ± 13% over 7 days in untreated rats; intetumumab increased tumor rCBV by 65 ± 10%, whereas bevacizumab reduced tumor rCBV by 31 ± 10% at 7 days (P < .001 for group and day). Similarly, intetumumab increased brain tumor vascular permeability compared with controls at 3 and 7 days after treatment, whereas bevacizumab decreased tumor permeability within 24 hours (P = .0004 for group, P = .0081 for day). All tumors grew over the 7-day assessment period, but bevacizumab slowed the increase in tumor volume on MRI. We conclude that the vascular targeting agents intetumumab and bevacizumab had diametrically opposite effects on dynamic MRI of tumor vasculature in rat brain tumor models. Targeting α(V)-integrins increased tumor vascular permeability and blood volume, whereas bevacizumab decreased both measures. These findings have implications for chemotherapy delivery and antitumor efficacy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Neovascularização Patológica/prevenção & controle , Inibidores da Angiogênese/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados , Bevacizumab , Neoplasias Encefálicas/tratamento farmacológico , Permeabilidade Capilar , Feminino , Gadolínio DTPA , Humanos , Integrina alfaV/química , Integrina alfaV/imunologia , Ratos , Ratos Nus , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/imunologia
8.
Cancer Res ; 70(19): 7591-9, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20841470

RESUMO

We previously reported that intetumumab (CNTO 95), a fully human anti-αv integrin monoclonal antibody, is a radiosensitizer in mice with xenograft tumors. Because intetumumab does not cross-react with mouse integrins, but has cross-reactivity with rat integrins, we next studied the potential combined use of radiation therapy and intetumumab in human cancer xenograft models in nude rats to assess effects on both tumor cells and the tumor microenvironment. Nude rats bearing human head and neck cancer and non-small cell lung cancer (NSCLC) xenografts were treated with intetumumab and fractionated local tumor radiotherapy. Effects on tumor growth and metastasis, blood perfusion, oxygenation, and gastrointestinal toxicity were studied. Intetumumab alone had a moderate effect on tumor growth. When combined with fractionated radiation therapy, intetumumab significantly inhibited tumor growth and produced a tumor response rate that was significantly better than with radiation therapy alone. Treatment with intetumumab also significantly reduced lung metastasis in the A549 NSCLC xenograft model. The oxygenation and blood perfusion in xenograft tumors measured by microbubble-enhanced ultrasound imaging were substantially increased after treatment with intetumumab. The combined use of intetumumab and radiation therapy reduced the microvessel density and increased apoptosis in tumor cells and the tumor microenvironment. Toxicity studies showed that treatment with intetumumab did not cause the histopathologic changes in the lungs and did not sensitize the sensitive gastrointestinal epithelium to the effect of radiation therapy. Intetumumab can potentiate the efficacy of fractionated radiation therapy in human cancer xenograft tumors in nude rats without increased toxicity.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Radiossensibilizantes/farmacologia , Animais , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma Pulmonar de Células não Pequenas/secundário , Linhagem Celular Tumoral , Terapia Combinada , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Integrina alfaV/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Mucosa Intestinal/efeitos da radiação , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/secundário , Masculino , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação , Oxigênio/sangue , Oxigênio/metabolismo , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Med Primatol ; 35(1): 12-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16430490

RESUMO

BACKGROUND: Prostate-specific antigen (PSA) is an invaluable tumor marker for the detection of early prostate cancer, and can be a target for active immunotherapy of prostate cancer. We wanted to assess the usefulness of the cynomolgus monkey (Macaca fascicularis) as a relevant animal model to evaluate PSA-specific therapies. METHODS: RNA was isolated from the prostate of cynomolgus monkeys, and PSA gene products were amplified by reverse transcriptase-polymerase chain reaction using primers from conserved regions of human and rhesus monkey (Macaca mulatta) PSA genes. These amplified products were then sequenced. RESULTS: The cynomolgus PSA amino acid sequence is 89.7% identical to the human PSA gene, and 99.2% identical to the rhesus PSA amino acid sequence. Like the human and rhesus PSA genes, an open-reading frame of 261 amino acids was identified for the cynomolgus gene. Expression of the cynomolgus PSA gene appears to be restricted to the prostate, as it is in humans. CONCLUSIONS: The high identity between human and cynomolgus PSA sequences suggests that the cynomolgus monkey should be studied further for its potential as a large animal model to evaluate PSA-specific therapies.


Assuntos
Macaca fascicularis/genética , Antígeno Prostático Específico/genética , Regiões 3' não Traduzidas/genética , Sequência de Aminoácidos , Animais , Composição de Bases , Clonagem Molecular , DNA Complementar/genética , Masculino , Dados de Sequência Molecular , Antígeno Prostático Específico/química , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie
10.
Vaccine ; 24(3): 244-53, 2006 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-16135392

RESUMO

DNA vaccines show efficacy in many preclinical models, but these results have not yet translated to consistent clinical efficacy. Co-administration of molecularly encoded adjuvants is one approach that may enable DNA vaccines to achieve enhanced immune response induction in humans. Interleukin-18 (IL-18) is a Th1-type cytokine that has been shown to augment the activity of DNA vaccines in some preclinical models. A prostate-specific antigen (PSA) DNA vaccine was tested in a mouse tumor model system to explore the impact of co-administration of a pIL-18 plasmid. Low doses of the pPSA vaccine were not capable of inducing tumor protection, but when pIL-18 was co-administered, complete tumor protection was observed in all mice. Tumor protection was mediated by both CD4(+) and CD8(+) T cells. Detailed analysis of the immune response in mice immunized with either pPSA or pPSA/pIL-18 demonstrated that pIL-18 skewed the PSA-specific immune response toward Th1. More importantly, stronger CD4(+) and CD8(+) T cell responses developed in the pPSA/pIL-18-immunized mice, with faster kinetics. These results suggest that IL-18 is a powerful adjuvant molecule that can enhance the development of antigen-specific immunity and vaccine efficacy.


Assuntos
Vacinas Anticâncer/imunologia , Interleucina-18/farmacologia , Células Th1/imunologia , Adjuvantes Imunológicos , Animais , Especificidade de Anticorpos , Antimetabólitos , Bromodesoxiuridina , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/análise , Citocinas/biossíntese , Feminino , Imunoglobulina G/análise , Imunoglobulina G/biossíntese , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Neoplasias/prevenção & controle , Plasmídeos/genética , Estimulação Química , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia
11.
Cancer Immunol Immunother ; 54(11): 1082-94, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16047142

RESUMO

Prostate specific antigen (PSA) is a serum marker that is widely used in the detection and monitoring of prostate cancer. Though PSA is a self-antigen, T cell responses to PSA epitopes have been detected in healthy men and prostate cancer patients, suggesting it may be used as a target for active immunotherapy of prostate cancer. A PSA DNA vaccine (pPSA) was evaluated in mice and monkeys for its ability to induce antigen-specific immune responses. Mice immunized intradermally with pPSA demonstrated strong PSA-specific humoral and cellular immunity. The anti-PSA immune responses were skewed toward Th1, as shown by high IFNgamma and IL-2 production. The immune response was sufficient to protect mice from challenge with PSA-expressing tumor cells. Tumor protection was durable in the absence of additional vaccination, as demonstrated by protection of vaccinated mice from tumor rechallenge. Furthermore, pPSA vaccination induced PSA-specific antibody titers in male cynomolgus monkeys, which express a closely related PSA gene. These results demonstrate that vaccination with pPSA may be able to break tolerance and can induce an immune response that mediates tumor protection.


Assuntos
Antígeno Prostático Específico/imunologia , Neoplasias da Próstata/prevenção & controle , Células Th1/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos/sangue , Feminino , Humanos , Imunização , Interferon gama/biossíntese , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA