Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Genet ; 10(4): 430-5, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7670493

RESUMO

We have achieved efficient transduction of tumour metastases in vivo by the vascular delivery of retroviral producer cells. Experimental liver metastases in mice were created by intrasplenic injection of tumour cells into the portal venous circulation. Following the establishment of micrometastases, delivery of retroviral producer cells by the same route with a vector containing the Escherichia coli beta-galactosidase (lacZ) gene demonstrated selective in vivo gene transfer to tumour deposits. By this approach, two retroviral producer cell lines encoding cytokines (IL-4 and IL-2) directed tumoricidal inflammatory responses to established metastases. Cytokine gene targeting inhibited metastasis formation and caused significant overall reduction in tumour burden. These results suggest a novel therapeutic approach for the treatment of disseminated cancer.


Assuntos
Marcação de Genes , Terapia Genética , Interleucina-2/genética , Interleucina-4/genética , Neoplasias Hepáticas/secundário , Retroviridae/genética , Células 3T3 , Animais , Feminino , Técnicas de Transferência de Genes , Interleucina-2/uso terapêutico , Interleucina-4/uso terapêutico , Óperon Lac , Neoplasias Hepáticas/terapia , Masculino , Camundongos , Células Tumorais Cultivadas
2.
Science ; 264(5159): 713-6, 1994 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-8171324

RESUMO

The in vivo function of murine granulocyte-macrophage colony-stimulating factor (GM-CSF) was investigated in mice, carrying a null allele of the GM-CSF gene, that were generated by gene targeting techniques in embryonic stem cells. Although steady-state hematopoiesis was unimpaired in homozygous mutant animals, all animals developed the progressive accumulation of surfactant lipids and proteins in the alveolar space, the defining characteristic of the idiopathic human disorder pulmonary alveolar proteinosis. Extensive lymphoid hyperplasia associated with lung airways and blood vessels was also found, yet no infectious agents could be detected. These results demonstrate that GM-CSF is not an essential growth factor for basal hematopoiesis and reveal an unexpected, critical role for GM-CSF in pulmonary homeostasis.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Pulmão/patologia , Proteinose Alveolar Pulmonar/patologia , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/química , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Hematopoese , Homeostase , Humanos , Hiperplasia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteolipídeos/metabolismo , Proteinose Alveolar Pulmonar/metabolismo , Alvéolos Pulmonares/patologia , Proteínas Associadas a Surfactantes Pulmonares
3.
J Clin Invest ; 97(3): 649-55, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8609219

RESUMO

Mutation of the granulocyte-macrophage colony-stimulating factor (GM-CSF) gene by homologous recombination caused alveolar proteinosis in mice. To further discern the role of GM-CSF in surfactant homeostasis, the synthesis of GM-CSF was directed to the respiratory epithelium of GM-CSF-hull mutant mice (GM-/-) with a chimeric gene expressing GM-CSF under the control of the promoter from the human surfactant protein-C (SP-C) gene. Transgenic mice bearing the SP-C-GM-CSF construct (SP-C-GM+) were bred to GM-/- mice resulting in complete correction of alveolar proteinosis in bitransgenic GM-/-, SP-C-GM+ mice. No effects of the transgene were found outside the lung. GM-CSF was increased in bronchoalveolar lavage fluid of the bitransgenic mice. Surfactant proteins-A and -B and phospholipid in bronchoalveolar lavage fluid were normalized in the GM-/-, SP-C-GM+ mice. SP-A, -B, and -C mRNAs were unaltered in lungs from GM-CSF-deficient and -replete mice. Expression of GM-CSF in respiratory epithelial cells of transgenic mice restores surfactant homeostasis in GM-/- mice. From these findings, we conclude that GM-CSF regulates the clearance or catabolism rather than synthesis of surfactant proteins and lipids.


Assuntos
Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Proteinose Alveolar Pulmonar/terapia , Animais , Modelos Animais de Doenças , Epitélio/metabolismo , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Pulmão/anatomia & histologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteolipídeos/genética , Proteinose Alveolar Pulmonar/genética , Surfactantes Pulmonares/genética , Surfactantes Pulmonares/metabolismo , Distribuição Tecidual
4.
J Clin Invest ; 106(5): 705-14, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10974024

RESUMO

The effectiveness of donor-lymphocyte infusion (DLI) for treatment of relapsed chronic myelogenous leukemia (CML) after allogeneic bone marrow transplantation is a clear demonstration of the graft-versus-leukemia (GVL) effect. T cells are critical mediators of GVL, but the antigenic targets of this response are unknown. To determine whether patients who respond to DLI also develop B-cell immunity to CML-associated antigens, we analyzed sera from three patients with relapsed CML who achieved a complete molecular remission after infusion of donor T cells. Sera from these individuals recognized 13 distinct gene products represented in a CML-derived cDNA library. Two proteins, Jkappa-recombination signal-binding protein (RBP-Jkappa) and related adhesion focal tyrosine kinase (RAFTK), were recognized by sera from three of 19 DLI responders. None of these antigens were recognized by sera from healthy donors or patients with chronic graft-versus-host disease. Four gene products were recognized by sera from CML patients treated with hydroxyurea and nine were detected by sera from CML patients who responded to IFN-alpha. Antibody titers specific for RAFTK, but not for RBP-Jkappa, were found to be temporally associated with the response to DLI. These results demonstrate that patients who respond to DLI generate potent antibody responses to CML-associated antigens, suggesting the development of coordinated T- and B-cell immunity. The characterization of B cell-defined antigens may help identify clinically relevant targets of the GVL response in vivo.


Assuntos
Anticorpos Antineoplásicos/sangue , Efeito Enxerto vs Leucemia , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Transfusão de Linfócitos , Proteínas Nucleares , Linfócitos B/imunologia , Proteínas de Ligação a DNA/imunologia , Quinase 2 de Adesão Focal , Biblioteca Gênica , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Dados de Sequência Molecular , Proteínas Tirosina Quinases/imunologia , Indução de Remissão , Análise de Sequência de DNA , Linfócitos T/imunologia
5.
J Clin Invest ; 100(11): 2757-65, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9389740

RESUMO

Multiple clinical trials have shown the efficacy of adoptively transferred allogeneic antigen-specific T cells for the treatment of viral infections and relapsed hematologic malignancies. In contrast, the therapeutic potential of autologous antigen-specific T cells has yet to be established since it has been technically difficult to generate sufficient numbers of these T cells, ex vivo. A major obstacle to the success of this objective derives from our inability to simply and rapidly isolate and/or expand large numbers of highly efficient antigen presenting cells (APCs) for repetitive stimulations of antigen-specific T cells in vitro. We show that autologous CD40-activated B cells represent a readily available source of highly efficient APC that appear to have several important advantages over other APCs for ex vivo T cell expansion including: (a) methodological simplicity necessary to generate continuously large numbers of APCs from just 50 cm3 of peripheral blood without loss of APC function; (b) capacity to induce high peak T cell proliferation and interferon-gamma production without IL-10 production; (c) ease in cryopreservation; and (d) markedly reduced cost. We, therefore, contend that CD40-activated B cells are an alternative source of highly efficient APCs with which to generate antigen-specific T cells ex vivo for autologous adoptive immunotherapy.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos B/imunologia , Antígenos CD40/imunologia , Linfócitos T/imunologia , Células 3T3 , Animais , Antígenos de Neoplasias/imunologia , Antígenos Virais/imunologia , Divisão Celular , Células Cultivadas , Análise Custo-Benefício , Testes Imunológicos de Citotoxicidade , Células Dendríticas/citologia , Células Dendríticas/imunologia , Humanos , Imunoterapia Adotiva , Vírus da Influenza A/imunologia , Interferon gama/biossíntese , Interleucina-10/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Camundongos , Monofenol Mono-Oxigenase/imunologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/imunologia , Fatores de Tempo , Células Tumorais Cultivadas , Proteínas da Matriz Viral/síntese química , Proteínas da Matriz Viral/imunologia
6.
Curr Opin Immunol ; 12(5): 571-5, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11007361

RESUMO

Modification of the tumor microenvironment with gene transfer techniques stimulates two immune mechanisms that effectuate tumor destruction. One involves improved tumor-antigen presentation for the development of specific cellular and humoral immunity. The second involves compromise of the tumor vasculature by soluble factors and leukocytes.


Assuntos
Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-12/metabolismo , Animais , Humanos
7.
Cancer Res ; 45(9): 4082-6, 1985 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-4028000

RESUMO

The human glioma-derived cell line D-54 MG and the human medulloblastoma-derived cell line TE-671 have been shown to be sensitive in culture to the pharmacological interference with glutamine metabolism by acivicin, 6-diazo-5-oxo-L-norleucine, and methionine sulfoximine. Using as a guide the multiple contributions of glutamine to the biosynthesis of proteins, purines, and pyrimidines, we now have identified six additional antimetabolites active against these lines in vitro at clinically relevant concentrations. The 50% growth-inhibitory levels of the drugs against D-54 MG in 6-day continuous exposure experiments were: L-asparaginase, 0.057 IU/ml; 5-fluorouracil, 0.5 micrograms/ml; 6-mercaptopurine, 0.8 micrograms/ml; actinomycin D, 0.0007 micrograms/ml; N-phosphonacetyl-L-aspartic acid, 2.3 micrograms/ml; and 5-azacytidine, 0.2 micrograms/ml (3-day exposure. The corresponding 50% growth-inhibitory values in TE-671 were: L-asparaginase, 0.54 IU/ml; 5-fluorouracil, 1.5 micrograms/ml; 6-mercaptopurine, 4.7 micrograms/ml; actinomycin D, 0.00044 micrograms/ml; N-phosphonacetyl-L-aspartic acid, 4.5 micrograms/ml; and 5-azacytidine, 0.49 micrograms/ml. Dipyridamole up to 10 micrograms/ml was inactive against both lines. The isobologram method was used to evaluate the effectiveness of several two-drug combinations which were biochemically designed. The sums of the optimal fractional inhibitory concentrations for the pairs were: acivicin plus L-asparaginase, 0.14; acivicin plus methionine sulfoximine, 0.40; 6-diazo-5-oxo-L-norleucine plus methionine sulfoximine, 0.60; acivicin plus 6-mercaptopurine, 1.0, all in TE-671; and acivicin plus 5-fluorouracil, 0.79, in D-54 MG. Our findings suggest that an antimetabolite regimen exploiting glutamine sensitivity might improve the chemotherapy of some human gliomas and medulloblastomas.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Glioma/tratamento farmacológico , Glutamina/metabolismo , Meduloblastoma/tratamento farmacológico , Linhagem Celular , Relação Dose-Resposta a Droga , Glioma/metabolismo , Glutamina/antagonistas & inibidores , Humanos , Meduloblastoma/metabolismo
8.
Cancer Res ; 61(1): 162-71, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11196155

RESUMO

Allogeneic bone marrow transplantation (BMT) is currently restricted to hematological malignancies because of a lack of antitumor activity against solid cancers. We have tested a novel treatment strategy to stimulate specific antitumor activity against a solid tumor after BMT by vaccination with irradiated tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF). Using the B16 melanoma model, we found that vaccination elicited potent antitumor activity in recipients of syngeneic BMT in a time-dependent fashion, and that immune reconstitution was critical for the development of antitumor activity. Vaccination did not stimulate antitumor immunity after allogeneic BMT because of the post-BMT immunodeficiency associated with graft-versus-host disease (GVHD). Remarkably, vaccination was effective in stimulating potent and long-lasting antitumor activity in recipients of T-cell-depleted (TCD) allogeneic bone marrow. Recipients of TCD bone marrow who showed significant immune reconstitution by 6 weeks after BMT developed B16-specific T-cell-cytotoxic, proliferative, and cytokine responses as a function of vaccination. T cells derived from donor stem cells were, therefore, able to recognize tumor antigens, although they remained tolerant to host histocompatibility antigens. These results demonstrate that GM-CSF-based tumor cell vaccines after allogeneic TCD BMT can stimulate potent antitumor effects without the induction of GVHD, and this strategy has important implications for the treatment of patients with solid malignancies.


Assuntos
Transplante de Medula Óssea/imunologia , Vacinas Anticâncer/imunologia , Efeito Enxerto vs Tumor/imunologia , Melanoma Experimental/imunologia , Linfócitos T/imunologia , Animais , Feminino , Doença Enxerto-Hospedeiro/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Antígenos de Histocompatibilidade/imunologia , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL
9.
Cancer Res ; 60(12): 3239-46, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10866317

RESUMO

Both granulocyte-macrophage colony-stimulating factor (GM-CSF) and flt3-ligand (FL) induce the development of dendritic cells (DCs). To compare the functional properties of DCs stimulated by these cytokines in vivo, we used retroviral-mediated gene transfer to generate murine tumor cells secreting high levels of each molecule. Injection of tumor cells expressing either GM-CSF or FL resulted in the dramatic increase of CD11c+ cells in the spleen and tumor infiltrate. However, vaccination with irradiated, GM-CSF-secreting tumor cells stimulated more potent antitumor immunity than vaccination with irradiated, FL-secreting tumor cells. The superior antitumor immunity elicited by GM-CSF involved a broad T cell cytokine response, in contrast to the limited Thl response elicited by FL. DCs generated by GM-CSF were CD8alpha- and expressed higher levels of B7-1 and CD1d than DCs cells generated by FL. Injection sites of metastatic melanoma patients vaccinated with irradiated, autologous tumor cells engineered to secrete GM-CSF demonstrated similar, dense infiltrates of DCs expressing high levels of B7-1. These findings reveal critical differences in the abilities of GM-CSF and FL to enhance the function of DCs in vivo and have important implications for the crafting of tumor vaccines.


Assuntos
Adjuvantes Imunológicos/metabolismo , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Animais , Antígenos CD1/metabolismo , Antígenos CD1d , Antígenos CD8/metabolismo , Vacinas Anticâncer , Células Dendríticas/metabolismo , Feminino , Citometria de Fluxo , Técnicas de Transferência de Genes , Humanos , Integrina alfaXbeta2/metabolismo , Interleucina-5/metabolismo , Melanoma/imunologia , Melanoma/terapia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Retroviridae/genética , Baço/imunologia , Linfócitos T/imunologia , Células Th1/imunologia , Fatores de Tempo , Células Tumorais Cultivadas
10.
Cancer Res ; 45(9): 4077-81, 1985 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2862994

RESUMO

Cellular supply of glutamine, an essential substrate for growth, is derived from extracellular fluid and de novo synthesis. We investigated the relative importance of these sources to the growth of six human anaplastic glioma- and one human medulloblastoma-derived permanent cell lines. Exogenous glutamine was limiting for the proliferation of glioma-derived lines D-54 MG, U-118 MG, and U-251 MG. In contrast, medulloblastoma-derived line TE-671 and glioma-derived lines U-373 MG, D-245 MG, and D-259 MG grew in the absence of supplemental glutamine. Two cell lines with contrasting glutamine requirements, D-54 MG and TE-671, were used to explore the pharmacological interference with glutamine metabolism. DL-alpha-Aminoadipic acid, a reported glutamic acid analogue with gliotoxic properties, significantly inhibited the growth of both lines. These effects were reversed by increasing glutamine, suggesting that the major action of DL-alpha-aminoadipic acid is as a glutamine antagonist. In contrast, the glutamine synthetase inhibitor delta-hydroxylysine demonstrated activity only against TE-671. Acivicin and 6-diazo-5-oxo-L-norleucine, glutamine analogues available for clinical use, reduced the proliferation of both cell lines at pharmacological concentrations. Methionine sulfoximine, a glutamine synthetase inhibitor previously used clinically, produced marked growth inhibition only against TE-671. These findings indicate that the synthesis and utilization of glutamine are potentially exploitable targets for the chemotherapy of some human gliomas and medulloblastomas.


Assuntos
Glioma/patologia , Glutamina/farmacologia , Meduloblastoma/patologia , Ácido 2-Aminoadípico/farmacologia , Células Cultivadas , Diazo-Oxo-Norleucina/farmacologia , Relação Dose-Resposta a Droga , Glutamato-Amônia Ligase/análise , Humanos , Hidroxilisina/farmacologia
11.
Cancer Res ; 53(10 Suppl): 2221-6, 1993 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8485707

RESUMO

Preclinical studies with murine tumor models have demonstrated that autologous tumor cell vaccines engineered to secrete certain cytokines in a paracrine fashion elicit systemic immune responses capable of eliminating small amounts of established tumor. These results have engendered much interest in developing this strategy for gene therapy of human cancer. The major limitation to creating genetically modified autologous human tumor vaccines is efficient gene transfer into primary tumor explants, since the majority of human tumors fail to proliferate in long-term culture. Using the retroviral vector MFG in conjunction with short-term culture techniques, we have achieved, in the absence of selection, a mean transduction efficiency of 60% in primary renal, ovarian, and pancreatic tumor explants, and we have developed an autologous granulocyte-macrophage colony-stimulating factor secreting tumor vaccine for clinical trials.


Assuntos
Adenocarcinoma/genética , Neoplasias/genética , Transfecção/métodos , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Feminino , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Retroviridae/genética , Transdução Genética/genética , Transfecção/genética , Células Tumorais Cultivadas
12.
Cancer Res ; 57(13): 2569-74, 1997 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9205055

RESUMO

Interactions between CD40 and its ligand, CD154 (CD40L, gp39), have been shown to play a central role in the regulation of humoral immunity. Recent evidence suggests that this ligand-receptor pair also plays an important role in the induction of cell-mediated immune responses, including those directed against viral pathogens, intracellular parasites, and alloantigens. The contribution of this ligand-receptor pair to the development of protective immunity against syngeneic tumors was evaluated by blocking the in vivo function of CD154 or by studying tumor resistance in mice genetically deficient in CD40 expression (CD40-/-). In the former case, anti-CD154 monoclonal antibody treatment inhibited the generation of protective immune responses after the administration of three potent tumor vaccines: irradiated MCA 105, MCA 105 admixed with Corynebacterium parvum adjuvant, and irradiated B16 melanoma cells transduced with the gene for granulocyte macrophage colony-stimulating factor. Confirmation of the role of CD40/CD154 interactions in tumor immunity was provided by the overt tumor susceptibility in CD40-deficient mice as compared to that in CD40+/+ mice. In this case, wild-type but not CD40-deficient mice could be readily protected against live TS/A tumor challenge by preimmunization with TS/A admixed with C. parvum. These findings suggest a critical role for CD40/CD154 interactions in the induction of cellular immunity by tumor vaccines and may have important implications for future approaches to cell-based cancer therapies.


Assuntos
Antígenos CD40/fisiologia , Vacinas Anticâncer/imunologia , Glicoproteínas de Membrana/fisiologia , Adjuvantes Imunológicos/fisiologia , Animais , Ligante de CD40 , Feminino , Imunidade Celular/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Taxa de Sobrevida , Células Tumorais Cultivadas
13.
Cancer Res ; 57(8): 1537-46, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9108457

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transduced, irradiated tumor vaccines induce potent, T-cell-mediated antitumor immune responses in preclinical models. We report the initial results of a Phase I trial evaluating this strategy for safety and the induction of immune responses in patients with metastatic renal cell carcinoma (RCC). Patients were treated in a randomized, double-blind dose-escalation study with equivalent doses of autologous, irradiated RCC vaccine cells with or without ex vivo human GM-CSF gene transfer. The replication-defective retroviral vector MFG was used for GM-CSF gene transfer. No dose-limiting toxicities were encountered in 16 fully evaluable patients. GM-CSF gene-transduced vaccines were equivalent in toxicity to nontransduced vaccines up to the feasible limits of autologous tumor vaccine yield. No evidence of autoimmune disease was observed. Biopsies of intradermal sites of injection with GM-CSF gene-transduced vaccines contained distinctive macrophage, dendritic cell, eosinophil, neutrophil, and T-cell infiltrates similar to those observed in preclinical models of efficacy. Histological analysis of delayed-type hypersensitivity responses in patients vaccinated with GM-CSF-transduced vaccines demonstrated an intense eosinophil infiltrate that was not observed in patients who received nontransduced vaccines. An objective partial response was observed in a patient treated with GM-CSF gene-transduced vaccine who displayed the largest delayed-type hypersensitivity conversion. No replication-competent retrovirus was detected in vaccinated patients. This Phase I study demonstrated the feasibility, safety, and bioactivity of an autologous GM-CSF gene-transduced tumor vaccine for RCC patients.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/terapia , Técnicas de Transferência de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias Renais/terapia , Adulto , Idoso , Vacinas Anticâncer/efeitos adversos , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/efeitos da radiação , Carcinoma de Células Renais/imunologia , Vírus Defeituosos/genética , Método Duplo-Cego , Toxidermias/etiologia , Toxidermias/imunologia , Toxidermias/patologia , Feminino , Vetores Genéticos/genética , Humanos , Hipersensibilidade Tardia/patologia , Neoplasias Renais/imunologia , Masculino , Pessoa de Meia-Idade , Vacinação/efeitos adversos
14.
J Clin Oncol ; 16(7): 2548-56, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9667277

RESUMO

Molecular genetics has spawned an impressive outpouring of insights into the biology of neoplastic transformation and the host-tumor relationship. This deeper understanding of cancer pathogenesis presents a rich opportunity to develop novel therapeutic agents with improved selectivity for cancer cells. One promising approach involves gene therapy, which is the introduction of genetic material into a patient's tissues with the intent to achieve therapeutic benefit. A number of gene transfer systems have been designed that enable the genetic modification of relevant target cells, albeit with varying strengths and limitations. Several strategies to exploit gene transfer as a tool to target specific molecular defects intrinsic to cancer cells, enhance tumor chemosensitivity, and augment tumor immunogenicity are under intensive investigation. A number of these approaches have entered initial clinical testing and already provide intriguing new information about the biology of cancer in patients. In this review, I will highlight the critical issues and controversies that underscore preclinical experiments in cancer gene therapy, discuss some of the preliminary findings from the first wave of clinical trials, and speculate about the prospects that cancer gene therapy will change the way that cancer medicine is practiced.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Oncologia/métodos , Neoplasias/genética , Ensaios Clínicos como Assunto , Humanos , Estados Unidos
15.
J Leukoc Biol ; 64(6): 810-6, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9850165

RESUMO

We test the hypothesis that the monocyte-macrophage colony-stimulating factor (CSF-1 or M-CSF) plays a major role in the inflammatory responses of Mphi by acting as a priming agent that heightens their responsiveness to secondary stimulation by other mediators. We previously reported that CSF-1 induced peritoneal Mphi (PMphi) to transcribe several genes including interleukin-6 (Il6) and granulocyte-macrophage colony-stimulating factor (Csfgm). It was reported that the Il6 and Csfgm genes were individually regulated by different pathways but it was not clear to what extent the two genes interacted during Mphi-mediated inflammatory responses. We now show that CSF-1 induces the release of bioactive GM-CSF from mouse resident PMphi. GM-CSF induces Il6 gene expression and synergizes with CSF-1 to induce the release of large amounts of IL-6. PMphi from C57BL/6J-Csfgm(null) mice were shown to release minimal IL-6 in response to CSF-1 and to express a much reduced response to the highly stimulatory combination of CSF-1 and lipopolysaccharide (LPS). Exogenous recombinant GM-CSF restored the IL-6 response of GM-CSF null PMphi to a great extent but not completely. As controls, three other recombinant proteins were tested but of these only tumor necrosis factor alpha (TNF-alpha) was shown to synergize with both CSF-1 and GM-CSF. Using PMphi from mice deficient in the expression of the Il6 gene, it was shown that they released two- to threefold more GM-CSF in response to CSF-1 than their control counterparts. However, an exogenous supply of recombinant IL-6 had no effect on GM-CSF release. The data indicate that the pathways regulating Il6 gene expression are under the control of a complex network of cytokine interactions involving at least CSF-1, GM-CSF, and TNF-alpha, with the added possibility that IL-6 may exert modulatory activity within this network.


Assuntos
Regulação da Expressão Gênica/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Interleucina-6/genética , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos Peritoneais/metabolismo , Animais , Células Cultivadas , Sinergismo Farmacológico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
16.
Leukemia ; 29(1): 218-29, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24935722

RESUMO

We evaluated a cocktail of HLA-A2-specific peptides including heteroclitic XBP1 US184-192 (YISPWILAV), heteroclitic XBP1 SP367-375 (YLFPQLISV), native CD138260-268 (GLVGLIFAV) and native CS1239-247 (SLFVLGLFL), for their ability to elicit multipeptide-specific cytotoxic T lymphocytes (MP-CTLs) using T cells from smoldering multiple myeloma (SMM) patients. Our results demonstrate that MP-CTLs generated from SMM patients' T cells show effective anti-MM responses including CD137 (4-1BB) upregulation, CTL proliferation, interferon-γ production and degranulation (CD107a) in an HLA-A2-restricted and peptide-specific manner. Phenotypically, we observed increased total CD3(+)CD8(+) T cells (>80%) and cellular activation (CD69(+)) within the memory SMM MP-CTL (CD45RO(+)/CD3(+)CD8(+)) subset after repeated multipeptide stimulation. Importantly, SMM patients could be categorized into distinct groups by their level of MP-CTL expansion and antitumor activity. In high responders, the effector memory (CCR7(-)CD45RO(+)/CD3(+)CD8(+)) T-cell subset was enriched, whereas the remaining responders' CTL contained a higher frequency of the terminal effector (CCR7(-)CD45RO(-)/CD3(+)CD8(+)) subset. These results suggest that this multipeptide cocktail has the potential to induce effective and durable memory MP-CTL in SMM patients. Therefore, our findings provide the rationale for clinical evaluation of a therapeutic vaccine to prevent or delay progression of SMM to active disease.


Assuntos
Proteínas de Ligação a DNA/imunologia , Epitopos/imunologia , Mieloma Múltiplo/imunologia , Peptídeos/imunologia , Sindecana-1/imunologia , Linfócitos T Citotóxicos/imunologia , Fatores de Transcrição/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Proliferação de Células , Progressão da Doença , Feminino , Humanos , Memória Imunológica , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Fatores de Transcrição de Fator Regulador X , Linfócitos T Citotóxicos/citologia , Proteína 1 de Ligação a X-Box
17.
Hum Gene Ther ; 8(9): 1065-72, 1997 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-9189764

RESUMO

We have developed an ex vivo gene therapy paradigm for the treatment of brain tumors using granulocyte-macrophage colony-stimulating factor (GM-CSF). Murine B16 melanoma cells were infected with MFG recombinant retrovirus containing the mouse GM-CSF cDNA. Subcutaneous vaccination of syngeneic mice with irradiated GM-CSF-secreting B16 melanoma cells was capable of completely protecting animals against subsequent intracranial B16 tumor inoculation, with up to 5 x 10(3) cells. Histologic evaluation revealed the presence of neutrophils, eosinophils, and lymphocytes, including CD4+, CD8+, and CD45R+ cells, in the intracerebral inoculation site, peaking 4 days after intracranial inoculation. In contrast, nonvaccinated animals or animals vaccinated with irradiated, nontransduced B16 cells succumbed to intracranial tumor within 3 weeks after inoculation. Treatment of established intracranial B16 melanoma tumors with subcutaneous injection of irradiated GM-CSF-secreting B16 cells significantly delayed death, as compared to injection of irradiated nontransduced B16 cells or no treatment. In addition, treatment of established intracerebral GL261 gliomas by vaccination with irradiated GM-CSF-secreting B16 cells mixed with irradiated, transduced, or nontransduced GL261 cells also extended survival. These B16/GL261 co-vaccinations also improved outcome and, in some cases, induced immunological memory that protected survivors from subsequent intracranial challenge with GL261 tumor cells. These findings indicate that peripheral vaccination with irradiated tumor cells in the presence of GM-CSF-producing cells can initiate a potent antitumor immune response against intracranial neoplasms.


Assuntos
Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Vacinação/métodos , Animais , Neoplasias Encefálicas/imunologia , Feminino , Glioma/genética , Glioma/patologia , Glioma/virologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Melanoma/genética , Melanoma/patologia , Melanoma/virologia , Camundongos , Camundongos Endogâmicos C57BL , Retroviridae/genética , Transdução Genética , Células Tumorais Cultivadas
18.
Cancer Gene Ther ; 4(6): 345-52, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9408604

RESUMO

Brain tumors have an immunoprivileged status which contributes to their refractoriness to treatment. In this study, immune rejection of GL261 glioma tumors in the mouse brain was achieved by subcutaneous vaccination with GM-CSF-transduced glioma cells. Cultured GL261 cells were transduced to secrete murine GM-CSF using a retrovirus vector, then irradiated, and injected subcutaneously into H-2 matched C57BL/6 mice. In prevaccination studies, the median survival time (MST) of animals vaccinated with 5 x 10(4) or 5 x 10(5) GM-CSF-transduced cells 7 days prior to intracranial injection of 10(6) nontransduced, nonirradiated GL261 cells was significantly prolonged by 45-50% compared with animals vaccinated in parallel with nontransduced, irradiated glioma cells. In treatment of established gliomas, the MST of animals, which were treated subcutaneously with 5 X 10(6) irradiated GM-CSF-transduced cells 3 days after intracranial injection of 2 x 10(4) nontransduced cells, was prolonged significantly by 36% compared with animals treated with the same number of nontransduced, irradiated cells or to sham-treated animals. In prevaccination studies, histology of brain tumors 4 days after intracranial tumor cell injection revealed infiltrates of CD8+ lymphocytes and eosinophils, the latter exclusively in animals vaccinated with GM-CSF-transduced cells, Thus, subcutaneous injection of irradiated GM-CSF-transduced glioma cells can induce a potent immune response to intracranial gliomas both as a vaccination against subsequent intracranial glioma cell implantation and for treatment of established intracranial glioma.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer , Terapia Genética/métodos , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/prevenção & controle , Linhagem Celular , Feminino , Vetores Genéticos , Glioma/patologia , Glioma/prevenção & controle , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/biossíntese , Retroviridae , Taxa de Sobrevida , Fatores de Tempo , Transfecção/métodos , Células Tumorais Cultivadas
19.
J Neuroimmunol ; 103(1): 16-25, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10674985

RESUMO

To evaluate the efficacy and toxicity of dendritic cell (DC) based therapy for intracerebral gliomas, we utilized a cell line derived from an astrocytoma that arose spontaneously in a VM/Dk mouse. This astrocytoma mirrors human gliomas phenotypically, morphologically and secretes transforming growth factor (TGF)-betas, immunosuppressive cytokines secreted by human gliomas. Systemic vaccination of mice with DCs pulsed with tumor homogenate followed by intracranial tumor challenge produced a > 160% increase in median survival (p = 0.016) compared with mice vaccinated with PBS or unpulsed DCs (p = 0.083). Fifty percent of mice treated with pulsed DCs survived long-term. Immunologic memory was demonstrated by survival of mice rechallenged with tumor. Both cell-mediated and humoral immunity was induced. On histological examination only focal areas of demyelination at the tumor implantation site were present. There was no evidence that autoimmune encephalomyelitis was induced by DC vaccination. Therefore, in a murine model, vaccination with DCs pulsed with glioma tumor homogenate is a safe and effective therapy against a syngeneic glioma located in the immunologically privileged central nervous system (CNS).


Assuntos
Células da Medula Óssea/imunologia , Neoplasias Encefálicas/imunologia , Células Dendríticas/imunologia , Glioma/imunologia , Animais , Neoplasias Encefálicas/terapia , Encefalomielite Autoimune Experimental/prevenção & controle , Glioma/terapia , Camundongos , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas , Vacinação
20.
Neurosurgery ; 41(6): 1365-72; discussion 1372-3, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9402588

RESUMO

OBJECTIVE: The promise of immunotherapies developed against brain tumors in animal models has not been realized in human clinical trials. This may be because of the routine use of rodent tumors artificially induced by chemicals or viruses that do not accurately portray the intrinsic qualities of spontaneously arising human tumors and that often fail to incorporate the role of immunosuppressants, such as transforming growth factor-beta, that are secreted by human gliomas. From an astrocytoma that arose spontaneously in inbred VM/Dk mice, we have characterized a highly tumorigenic spontaneous murine astrocytoma cell line (SMA-560) that retains features of glial differentiation and naturally produces high levels of biologically active transforming growth factor-beta. We have used this model to determine whether cytokine production by tumor cells will inhibit intracerebral astrocytoma growth. METHODS: Packaging cell lines producing replication-incompetent retroviral vectors were used to transfect the SMA-560 cell line in vitro with the genes encoding the murine cytokines interleukin (IL)-2, IL-3, IL-4, IL-6, tumor necrosis factor-alpha, gamma-interferon, or granulocyte-macrophage colony-stimulating factor or the costimulatory molecule B7.1 (CD80). RESULTS: Mice challenged intracerebrally with 5000 untransfected SMA-560 cells all succumbed to tumor within 30 days, with a median survival of 25 days. In contrast, mice challenged with SMA-560 cells producing IL-2, IL-4, or tumor necrosis factor-alpha each had a more than 400% increase in median survival (P < 0.0001). In these groups, 78.3% (18 of 23 mice), 66.7% (10 of 15 mice), and 60% (6 of 10 mice) of the mice, respectively, remained alive without evidence of tumor for longer than 100 days after the initial tumor challenge. All other cytokines tested and the expression of B7.1 failed to result in an increase in median survival. CONCLUSION: Using a spontaneous astrocytoma model in an inbred mouse strain, we have shown that cytokine production by glial tumors can abrogate their tumorigenicity in vivo despite production of transforming growth factor-beta. These results predict that approaches directed at cytokine production within intracerebral astrocytomas may be efficacious in human trials and that the "immunological privilege" of the brain may not be absolute under such conditions.


Assuntos
Astrocitoma/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Citocinas/metabolismo , Animais , Testes de Carcinogenicidade , Feminino , Vetores Genéticos , Antígenos de Histocompatibilidade Classe I/análise , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos , Retroviridae/genética , Transfecção , Fator de Crescimento Transformador beta/biossíntese , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA