Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Mol Med (Berl) ; 99(5): 663-671, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33398468

RESUMO

Mesenchymal stem cells (MSCs) are promising candidates for the development of cell-based drug delivery systems for autoimmune inflammatory diseases, such as multiple sclerosis (MS). Here, we investigated the effect of Ro-31-8425, an ATP-competitive kinase inhibitor, on the therapeutic properties of MSCs. Upon a simple pretreatment procedure, MSCs spontaneously took up and then gradually released significant amounts of Ro-31-8425. Ro-31-8425 (free or released by MSCs) suppressed the proliferation of CD4+ T cells in vitro following polyclonal and antigen-specific stimulation. Systemic administration of Ro-31-8425-loaded MSCs ameliorated the clinical course of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, displaying a stronger suppressive effect on EAE than control MSCs or free Ro-31-8425. Ro-31-8425-MSC administration resulted in sustained levels of Ro-31-8425 in the serum of EAE mice, modulating immune cell trafficking and the autoimmune response during EAE. Collectively, these results identify MSC-based drug delivery as a potential therapeutic strategy for the treatment of autoimmune diseases. KEY MESSAGES: MSCs can spontaneously take up the ATP-competitive kinase inhibitor Ro-31-8425. Ro-31-8425-loaded MSCs gradually release Ro-31-8425 and exhibit sustained suppression of T cells. Ro-31-8425-loaded MSCs have more sustained serum levels of Ro-31-8425 than free Ro-31-8425. Ro-31-8425-loaded MSCs are more effective than MSCs and free Ro-31-8425 for EAE therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Indóis/administração & dosagem , Maleimidas/administração & dosagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Esclerose Múltipla/tratamento farmacológico , Transplante Heterólogo/métodos , Animais , Proliferação de Células/efeitos dos fármacos , Liberação Controlada de Fármacos , Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/imunologia , Inibidores Enzimáticos/sangue , Feminino , Humanos , Imunidade/efeitos dos fármacos , Indóis/sangue , Maleimidas/sangue , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/sangue , Esclerose Múltipla/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Distribuição Tecidual , Resultado do Tratamento
2.
J Neuroimmunol ; 285: 4-12, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26198912

RESUMO

Alemtuzumab, a monoclonal antibody directed against human CD52, is used in the treatment of MS. To characterize the impact of anti-CD52 administration, a monoclonal antibody to mouse CD52 (anti-muCD52) was generated and evaluated in EAE mouse models of MS. A single course of anti-muCD52 provided a therapeutic benefit accompanied by a reduction in the frequency of autoreactive T lymphocytes and production of pro-inflammatory cytokines. Examination of the CNS revealed a decrease in infiltrating lymphocytes, demyelination and axonal loss. Electrophysiological assessment showed preservation of axonal conductance in the spinal cord. These findings suggest that anti-CD52 therapy may help preserve CNS integrity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Glicoproteínas/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Axônios/efeitos dos fármacos , Axônios/imunologia , Axônios/patologia , Antígeno CD52 , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/patologia , Glicoproteínas/antagonistas & inibidores , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular
3.
Immunology ; 141(1): 123-31, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24116901

RESUMO

Alemtuzumab is a humanized monoclonal antibody specific for the CD52 protein present at high levels on the surface of B and T lymphocytes. In clinical trials, alemtuzumab has shown a clinical benefit superior to that of interferon-ß in relapsing-remitting multiple sclerosis patients. Treatment with alemtuzumab leads to the depletion of circulating lymphocytes followed by a repopulation process characterized by alterations in the number, proportions and properties of lymphocyte subsets. Of particular interest, an increase in the percentage of T cells with a regulatory phenotype (Treg cells) has been observed in multiple sclerosis patients after alemtuzumab. Since Treg cells play an important role in the control of autoimmune responses, the effect of alemtuzumab on Treg cells was further studied in vitro. Alemtuzumab effectively mediated complement-dependent cytolysis of human T lymphocytes and the remaining population was enriched in T cells with a regulatory phenotype. The alemtuzumab-exposed T cells displayed functional regulatory characteristics including anergy to stimulation with allogeneic dendritic cells and ability to suppress the allogeneic response of autologous T cells. Consistent with the observed increase in Treg cell frequency, the CD25(hi) T-cell population was necessary for the suppressive activity of alemtuzumab-exposed T cells. The mechanism of this suppression was found to be dependent on both cell-cell contact and interleukin-2 consumption. These findings suggest that an alemtuzumab-mediated increase in the proportion of Treg cells may play a role in promoting the long-term efficacy of alemtuzumab in patients with multiple sclerosis.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Esclerose Múltipla/imunologia , Linfócitos T Reguladores/imunologia , Alemtuzumab , Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Antígeno CD52 , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/imunologia , Sobrevivência Celular , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Glicoproteínas/imunologia , Humanos , Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Linfócitos T Reguladores/patologia
4.
J Neuroimmunol ; 261(1-2): 29-36, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23759318

RESUMO

Alemtuzumab is a monoclonal antibody against the CD52 antigen present at high levels on the surface of lymphocytes. While treatment of multiple sclerosis patients with alemtuzumab results in marked depletion of lymphocytes from the circulation, it has not been associated with a high incidence of serious infections. In a human CD52 transgenic mouse, alemtuzumab treatment showed minimal impact on the number and function of innate immune cells. A transient decrease in primary adaptive immune responses was observed post-alemtuzumab but there was little effect on memory responses. These results potentially help explain the level of immunocompetence observed in alemtuzumab-treated MS patients.


Assuntos
Imunidade Adaptativa/genética , Anticorpos Monoclonais Humanizados/fisiologia , Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Glicoproteínas/imunologia , Alemtuzumab , Animais , Antígenos CD/genética , Antígenos de Neoplasias/genética , Linfócitos B/imunologia , Antígeno CD52 , Células Cultivadas , Glicoproteínas/genética , Humanos , Imunidade Inata/genética , Camundongos , Camundongos Transgênicos , Linfócitos T/imunologia , Resultado do Tratamento
5.
Leuk Lymphoma ; 53(1): 130-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21740294

RESUMO

The interaction between CXCR4 on the surface of tumor cells and CXCL12 in the stroma is believed to contribute to tumor cell survival and protection against drug treatment. Inhibition of stromal survival signals by CXCR4 antagonists has been reported to render tumor cells more sensitive to chemotherapy, but little is known about potential synergy with monoclonal antibodies. In this study, administration of the small molecule CXCR4 antagonists plerixafor and GENZ-644494 was found to enhance the anti-tumor activity of the monoclonal antibodies alemtuzumab and rituximab in disseminated lymphoma models. The observed enhancement in therapeutic efficacy by CXCR4 antagonists appeared to involve several factors, including interference with the tumor-promoting signals delivered by CXCL12, disruption of the tumor/stroma interaction and mobilization of effector neutrophils capable of mediating antibody-dependent cell-mediated cytotoxicity. The involvement of neutrophils was further supported by the observed reversal in therapeutic benefit upon neutrophil depletion.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfoma/tratamento farmacológico , Receptores CXCR4/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Alemtuzumab , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Benzilaminas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Quimiotaxia/efeitos dos fármacos , Ciclamos , Sinergismo Farmacológico , Citometria de Fluxo , Compostos Heterocíclicos/administração & dosagem , Compostos Heterocíclicos/farmacologia , Humanos , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos SCID , Fosforilação/efeitos dos fármacos , Piridinas/administração & dosagem , Piridinas/farmacologia , Receptores CXCR4/metabolismo , Rituximab , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos
6.
Leuk Lymphoma ; 51(7): 1293-304, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20377308

RESUMO

Alemtuzumab is a recombinant humanized IgG1 monoclonal antibody directed against CD52, an antigen expressed on the surface of normal and malignant B and T lymphocytes. Alemtuzumab is approved for the treatment of B-cell chronic lymphocytic leukemia (B-CLL), but the exact mechanism by which the antibody depletes malignant lymphocytes in vivo is not clearly defined. To address this issue, the anti-tumor activity of alemtuzumab was studied in disseminated and subcutaneous xenograft tumor models. The density of CD52 target antigen on the surface of tumor cells appeared to correlate with the anti-tumor activity of alemtuzumab. Deglycosylation of alemtuzumab resulted in a loss of cytotoxicity in vitro and was found to abolish anti-tumor activity in vivo. Individual inactivation of effector mechanisms in tumor-bearing mice indicated that the protective activity of alemtuzumab in vivo was primarily dependent on ADCC mediated by neutrophils and to a lesser extent NK cells. Increasing the number of circulating neutrophils by treatment with G-CSF enhanced the anti-tumor activity of the antibody, thus providing further evidence for the involvement of neutrophils as effector cells in the activity of alemtuzumab.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Células Matadoras Naturais/imunologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Neutrófilos/imunologia , Alemtuzumab , Animais , Anticorpos Monoclonais Humanizados , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Antígeno CD52 , Células CHO , Cricetinae , Cricetulus , Feminino , Glicoproteínas/metabolismo , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Técnicas Imunoenzimáticas , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/patologia , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos SCID , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Immunology ; 128(2): 260-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19740383

RESUMO

Alemtuzumab is a humanized monoclonal antibody against CD52, an antigen found on the surface of normal and malignant lymphocytes. It is approved for the treatment of B-cell chronic lymphocytic leukaemia and is undergoing Phase III clinical trials for the treatment of multiple sclerosis. The exact mechanism by which alemtuzumab mediates its biological effects in vivo is not clearly defined and mechanism of action studies have been hampered by the lack of cross-reactivity between human and mouse CD52. To address this issue, a transgenic mouse expressing human CD52 (hCD52) was created. Transgenic mice did not display any phenotypic abnormalities and were able to mount normal immune responses. The tissue distribution of hCD52 and the level of expression by various immune cell populations were comparable to those seen in humans. Treatment with alemtuzumab replicated the transient increase in serum cytokines and depletion of peripheral blood lymphocytes observed in humans. Lymphocyte depletion was not as profound in lymphoid organs, providing a possible explanation for the relatively low incidence of infection in alemtuzumab-treated patients. Interestingly, both lymphocyte depletion and cytokine induction by alemtuzumab were largely independent of complement and appeared to be mediated by neutrophils and natural killer cells because removal of these populations with antibodies to Gr-1 or asialo-GM-1, respectively, strongly inhibited the activity of alemtuzumab whereas removal of complement by treatment with cobra venom factor had no impact. The hCD52 transgenic mouse appears to be a useful model and has provided evidence for the previously uncharacterized involvement of neutrophils in the activity of alemtuzumab.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Antineoplásicos/imunologia , Glicoproteínas/metabolismo , Depleção Linfocítica/métodos , Modelos Animais , Adenoviridae/imunologia , Alemtuzumab , Animais , Anticorpos Monoclonais Humanizados , Anticorpos Antivirais/biossíntese , Antígeno CD52 , Citocinas/biossíntese , Citocinas/sangue , Relação Dose-Resposta Imunológica , Humanos , Tecido Linfoide/imunologia , Camundongos , Camundongos Transgênicos , Neutrófilos/imunologia , Baço/imunologia , Distribuição Tecidual
8.
Clin Transl Sci ; 2(1): 75-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20443871

RESUMO

Immunization with the electrofusion product of tumor cells and dendritic cells (DCs) is a promising approach to cancer immunotherapy. Production of electrofusion vaccines currently requires the acquisition of tumor material and must be tailored to each individual. Alternative vaccine configurations were explored in this study. Results indicated that fusion vaccines with fully syngeneic, semi-allogeneic or fully allogeneic components, were all effective in inducing specific, long-lasting antitumor immunity. This previously undescribed activity of a fully allogeneic fusion product introduces the possibility of using defined allogeneic tumor and DC lines to simplify vaccine manufacturing.


Assuntos
Fusão Celular/métodos , Células Dendríticas/imunologia , Eletroporação/métodos , Imunidade/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Apresentação Cruzada/imunologia , Citocinas/metabolismo , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Transplante Homólogo
9.
Hum Gene Ther ; 20(1): 11-20, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18828728

RESUMO

The use of adeno-associated viral (AAV) vectors for gene replacement therapy is currently being explored in several clinical indications. However, reports have suggested that input capsid proteins from AAV-2 vector particles may result in the stimulation of cytotoxic T lymphocyte (CTL) responses that can result in a loss of transduced cells. To explore the impact of anti-AAV CTLs on AAV-mediated transgene expression, both immunocompetent C57BL=6 mice and B cell-deficient muMT mice were immunized against the AAV2 capsid protein (Cap) and were injected intravenously with an AAV-2 vector encoding alpha-galactosidase (alpha-Gal). C57BL=6 mice, which developed both CTL and neutralizing antibody responses against Cap, failed to show any detectable alpha-Gal expression. In contrast, serum alpha-Gal levels comparable to those of naive mice were observed in muMT mice despite the presence of robust CTL activity against Cap, indicating that preexisting Cap-specific CTLs did not have any effect on the magnitude and duration of transgene expression. The same strategy was used to assess the impact of CTLs against the alpha-Gal transgene product on AAV-mediated gene delivery and persistence of transgene expression. Preimmunization of muMT mice with an Ad=alpha-Gal vector induced a robust CTL response to alpha-Gal. When these mice were injected with AAV2=alpha-Gal vector, initial levels of alpha-Gal expression were reduced by more than 1 log and became undetectable by 2 weeks postinjection. Overall, our results indicate that CTLs against the transgene product as opposed to AAV capsid protein are more likely to interfere with AAV transgene expression.


Assuntos
Proteínas do Capsídeo/imunologia , Dependovirus/metabolismo , Linfócitos T Citotóxicos/imunologia , Transgenes/imunologia , alfa-Galactosidase/metabolismo , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Dependovirus/genética , Dependovirus/imunologia , Terapia Genética , Vetores Genéticos , Imunocompetência , Hospedeiro Imunocomprometido , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptídeos/química , Peptídeos/genética , Peptídeos/imunologia , Peptídeos/metabolismo , Transgenes/genética , Transgenes/fisiologia , alfa-Galactosidase/genética , alfa-Galactosidase/imunologia
10.
Nutrition ; 23(3): 187-95, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17291720

RESUMO

OBJECTIVE: This study tested the hypothesis that supplementation of chromium picolinate (CrPic), 200 microg Cr/d, compared with an equivalent amount of picolinic acid (1720 microg) in CrPic and placebo, decreases body weight, alters body composition, and reduces iron status of women fed diets of constant energy and nutrients. METHODS: We fed 83 women nutritionally balanced diets, used anthropometry and dual x-ray absorptiometry to assess body composition, and measured serum and urinary Cr and biochemical indicators of iron status before and serially every 4 wk for 12 wk in a double-blind, randomized trial. RESULTS: CrPic supplementation increased (P < 0.0001) serum Cr concentration and urinary Cr excretion compared with picolinic acid and placebo. CrPic did not affect body weight or fat, although all groups lost (P < 0.05) weight and fat; it did not affect fat-free, mineral-free mass or measurements of iron status. CONCLUSION: Under conditions of controlled energy intake, CrPic supplementation of women did not independently influence body weight or composition or iron status. Thus, claims that supplementation of 200 microg of Cr as CrPic promotes weight loss and body composition changes are not supported.


Assuntos
Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Quelantes de Ferro/farmacologia , Ferro/sangue , Ácidos Picolínicos/farmacologia , Absorciometria de Fóton , Adulto , Antropometria , Biomarcadores/sangue , Biomarcadores/urina , Cromo/sangue , Cromo/farmacologia , Cromo/urina , Suplementos Nutricionais , Método Duplo-Cego , Feminino , Humanos , Pessoa de Meia-Idade , Obesidade/tratamento farmacológico , Ácidos Picolínicos/sangue , Ácidos Picolínicos/urina , Oligoelementos/sangue , Oligoelementos/farmacologia , Oligoelementos/urina , Resultado do Tratamento , Redução de Peso
11.
Mol Ther ; 7(4): 498-505, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12727113

RESUMO

Dendritic cells (DCs) are potent antigen-presenting cells capable of inducing primary T-cell responses. Several immunotherapy treatment strategies involve manipulation of DCs, both in vivo and ex vivo, to promote the immunogenic presentation of tumor-associated antigens. In this study, an electrofusion protocol was developed to induce fusion between tumor cells and allogeneic bone marrow-derived DCs. Preimmunization with irradiated electrofusion product was found to provide partial to complete protection from tumor challenge in the murine Renca renal cell carcinoma model and the B16 and M3 melanoma models. Vaccinated survivors developed specific immunological memory and were able to reject a subsequent rechallenge with the same tumor cells but not a syngeneic unrelated tumor line. Antitumor protection in the B16 model was accompanied by the development of a polyclonal cytotoxic T-lymphocyte response against defined melanoma-associated antigens. The therapeutic potential of this type of approach was suggested by the ability of a Renca-DC electrofusion product to induce tumor rejection in a substantial percentage of hosts (60%) bearing pre-established tumor cells. These results indicate that treatment with electrofused tumor cells and allogeneic DCs is capable of inducing a potent antitumor response and could conceivably be applied to a wide range of cancer indications for which tumor-associated antigens have not been identified.


Assuntos
Vacinas Anticâncer , Fusão Celular/métodos , Células Dendríticas/imunologia , Eletroporação , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/prevenção & controle , Animais , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/imunologia , Células Dendríticas/metabolismo , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
12.
Mol Ther ; 6(4): 519-27, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12377194

RESUMO

We have demonstrated recently that treatment of established peritoneal mesothelial tumors with complexes composed of cationic lipid and noncoding plasmid DNA (pNull) results in the inhibition of tumor growth accompanied by the induction of a tumor-specific cellular immune response. In this study, treatment of mice bearing intraperitoneal (i.p.) M3 melanoma tumors with i.p. injections of lipid/pNull complex was found to inhibit tumor growth and induce the development of a cytolytic response against several M3 melanoma-associated antigens. Depletion of CD8(+) T cells, as opposed to natural killer (NK) or CD4(+) T cells, essentially abrogated the therapeutic effect of lipid+pNull complex, thus supporting the involvement of cytotoxic CD8(+) T cells in the antitumor response. The antitumor effect of lipid/pNull complex was maximal following delivery into a tumor-bearing compartment. For example, i.p. delivery of complex was more effective than intravenous (i.v.) or subcutaneous (s.c.) treatment of i.p. M3 tumors. In addition, i.v. injection of complex displayed therapeutic activity against lung metastases caused by i.v. injection of tumor cells, and intratumoral injection of complex into solid s.c. tumors caused regression in most animals. Importantly, the immune response induced by local treatment of tumors with complex also offered systemic protection against tumor cells at distal sites, as illustrated by the eradication of both peritoneal tumors and lung metastases in mice treated with complex delivered i.p. Treatment with lipid/pNull complex, therefore, represents an attractive immune-based treatment modality that could potentially be applied to many tumor types.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica/genética , Terapia Genética , Melanoma Experimental/terapia , Plasmídeos , Linfócitos T Citotóxicos/imunologia , Animais , Portadores de Fármacos , Lipossomos , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Plasmídeos/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA