Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
2.
Cancer Immunol Immunother ; 60(1): 15-22, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21080166

RESUMO

Many assays to evaluate the nature, breadth, and quality of antigen-specific T cell responses are currently applied in human medicine. In most cases, assay-related protocols are developed on an individual laboratory basis, resulting in a large number of different protocols being applied worldwide. Together with the inherent complexity of cellular assays, this leads to unnecessary limitations in the ability to compare results generated across institutions. Over the past few years a number of critical assay parameters have been identified which influence test performance irrespective of protocol, material, and reagents used. Describing these critical factors as an integral part of any published report will both facilitate the comparison of data generated across institutions and lead to improvements in the assays themselves. To this end, the Minimal Information About T Cell Assays (MIATA) project was initiated. The objective of MIATA is to achieve a broad consensus on which T cell assay parameters should be reported in scientific publications and to propose a mechanism for reporting these in a systematic manner. To add maximum value for the scientific community, a step-wise, open, and field-spanning approach has been taken to achieve technical precision, user-friendliness, adequate incorporation of concerns, and high acceptance among peers. Here, we describe the past, present, and future perspectives of the MIATA project. We suggest that the approach taken can be generically applied to projects in which a broad consensus has to be reached among scientists working in fragmented fields, such as immunology. An additional objective of this undertaking is to engage the broader scientific community to comment on MIATA and to become an active participant in the project.


Assuntos
Consenso , Neoplasias/imunologia , Linfócitos T/imunologia , Alergia e Imunologia/tendências , Humanos , Técnicas Imunológicas/normas , Monitorização Fisiológica/normas , Guias de Prática Clínica como Assunto , Desenvolvimento de Programas , Projetos de Pesquisa
3.
Nat Med ; 5(7): 780-7, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10395323

RESUMO

Tumor antigen-specific T-cell tolerance limits the efficacy of therapeutic cancer vaccines. Antigen-presenting cells mediate the induction of T-cell tolerance to self-antigens. We therefore assessed the fate of tumor-specific CD4+ T cells in tumor-bearing recipients after in vivo activation of antigen-presenting cells with antibodies against CD40. Such treatment not only preserved the responsiveness of this population, but resulted in their endogenous activation. Established tumors regressed in vaccinated mice treated with antibody against CD40 at a time when no response was achieved with vaccination alone. These results indicate that modulation of antigen-presenting cells may be a useful strategy for enhancing responsiveness to immunization.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Vacinas Anticâncer , Carcinoma de Células Renais/prevenção & controle , Neoplasias Renais/prevenção & controle , Neoplasias Pulmonares/secundário , Glicoproteínas de Membrana/imunologia , Transferência Adotiva , Animais , Ligante de CD40 , Carcinoma de Células Renais/imunologia , Tolerância Imunológica , Neoplasias Renais/imunologia , Neoplasias Pulmonares/prevenção & controle , Transfusão de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Células Tumorais Cultivadas
4.
J Exp Med ; 179(4): 1215-24, 1994 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-7908321

RESUMO

Downregulation of major histocompatibility complex (MHC) class I expression is an important mechanism by which tumors evade classical T cell-dependent immune responses. Therefore, a system was designed to evaluate parameters for active immunization against MHC class I- tumors. Mice were capable of rejecting a MHC class I- tumor challenge after immunization with an irradiated granulocyte/macrophage colony-stimulating factor (GM-CSF) transduced MHC class I- tumor vaccine. This response was critically dependent on CD4+ T cells and natural killer (NK) cells, but minimally on CD8+ T cells. A strong protective response against MHC class I+ variants of the tumor could be elicited when mice were immunized with irradiated MHC class I+ GM-CSF-secreting tumor cells. This response required CD4+ and CD8+ T cells, and in addition, elimination of NK cells resulted in outgrowth of tumors that had lost expression of at least one MHC class I gene. Finally, class I MHC expression on the vaccinating cells inhibited the response generated against a MHC class I- tumor challenge. These results demonstrate that the host is capable of being immunized against a tumor that has lost MHC class I expression and reveal conditions under which distinct effector cells play a role in the systemic antitumor immune response.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Neoplasias Experimentais/imunologia , Linfócitos T/imunologia , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , DNA , Regulação para Baixo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Transplante de Neoplasias , Linfócitos T Reguladores/imunologia , Transdução Genética , Células Tumorais Cultivadas , Vacinação
5.
J Exp Med ; 182(5): 1415-21, 1995 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7595212

RESUMO

Introduction of the B7-1 gene into murine tumor cells can result in rejection of the B7-1 transductants and, in some cases, systemic immunity to subsequent challenge with the nontransduced tumor cells. These effects have been largely attributed to the function of B7-1 as a costimulator in directly activating tumor specific, major histocompatibility class I-restricted CD8+ T cells. We examined the role of B7-1 expression in the direct rejection as well as in the induction of systemic immunity to a nonimmunogenic murine tumor. B-16 melanoma cells with high levels of B7-1 expression did not grow in C57BL/6 recipient mice, while wild-type B-16 cells and cells with low B7-1 expression grew progressively within 21 d. In mixing experiments with B7-1hi and wild-type B-16 cells, tumors grew out in vivo even when a minority of cells were B7-1-. Furthermore, the occasional tumors that grew out after injection of 100% B-16 B7-1hi cells showed markedly decreased B7-1 expression. In vivo antibody depletions showed that NK1.1 and CD8+ T cells, but not CD4+ T cells, were essential for the in vivo rejection of tumors. Animals that rejected B-16 B7-1hi tumors did not develop enhanced systemic immunity against challenge with wild-type B-16 cells. These results suggest that a major role of B7-1 expression by tumors is to mediate direct recognition and killing by natural killer cells. With an intrinsically nonimmunogenic tumor, this direct killing does not lead to enhanced systemic immunity.


Assuntos
Antígeno B7-1/imunologia , Regulação Neoplásica da Expressão Gênica , Rejeição de Enxerto/imunologia , Subpopulações de Linfócitos/imunologia , Melanoma Experimental/imunologia , Proteínas de Neoplasias/imunologia , Transplante de Neoplasias/imunologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Citotoxicidade Imunológica , Imunidade Celular , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Recombinantes de Fusão/imunologia , Transfecção , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/transplante
6.
J Exp Med ; 191(3): 541-50, 2000 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-10662799

RESUMO

In many cases, induction of CD8(+) CTL responses requires CD4(+) T cell help. Recently, it has been shown that a dominant pathway of CD4(+) help is via antigen-presenting cell (APC) activation through engagement of CD40 by CD40 ligand on CD4(+) T cells. To further study this three cell interaction, we established an in vitro system using dendritic cells (DCs) as APCs and influenza hemagglutinin (HA) class I and II peptide-specific T cell antigen receptor transgenic T cells as cytotoxic T lymphocyte precursors and CD4(+) T helper cells, respectively. We found that CD4(+) T cells can provide potent help for DCs to activate CD8(+) T cells when antigen is provided in the form of either cell lysate, recombinant protein, or synthetic peptides. Surprisingly, this help is completely independent of CD40. Moreover, CD40-independent CD4(+) help can be documented in vivo. Finally, we show that CD40-independent T cell help is delivered through both sensitization of DCs and direct CD4(+)-CD8(+) T cell communication via lymphokines. Therefore, we conclude that CD4(+) help comprises at least three components: CD40-dependent DC sensitization, CD40-independent DC sensitization, and direct lymphokine-dependent CD4(+)-CD8(+) T cell communication.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T , Comunicação Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Ativação Linfocitária , Linfocinas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais , Linfócitos T Citotóxicos/imunologia
7.
J Exp Med ; 183(3): 769-76, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8642281

RESUMO

Tumors engineered to express the costimulatory molecule B7-1 can elicit CD8+ cytotoxic T lymphocyte (CTL)-dependent antitumor responses in immunocompetent mice. It has been postulated that this result reflects direct priming of CTL by the modified tumor in vivo. Previous studies of the immune response to a B7-1- murine colon carcinoma expressing influenza nucleoprotein (NP) as a model tumor antigen have demonstrated the crucial role of bone marrow-derived antigen-presenting cells (APCs) in the priming of NP-specific CTL in vivo. In this system, no evidence of direct CTL priming by tumor was detected. We have performed a similar analysis to determine if B7-1 transfectant of this tumor results in the direct priming of CTL, and to compare this response to that primed by host APCs. When H-2b-->H-2bxd bone marrow chimeras were immunized with a single injection of CT26/NP/B7-1 (H-2d), NP-specific CTL were detected that were restricted to the bone marrow haplotype (H-2b), but not to the tumor haplotype. In contrast, CTL recognizing the NP antigenic epitope in the context of the tumor's major histocompatibility complex were detectable only after multiple immunizations. These results suggest that whereas B7-1+ tumor vaccines result in some degree of direct presentation to CD8+ T cells, the dominant mechanism of CTL priming is through the uptake and presentation of tumor antigens by bone marrow-deprived APCs. However, repeated immunization with B7-1+ tumor cells can efficiently expand the directly primed CD8+ CTL population.


Assuntos
Antígeno B7-1/biossíntese , Capsídeo/imunologia , Neoplasias do Colo/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas do Core Viral/imunologia , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Medula Óssea/imunologia , Capsídeo/biossíntese , Capsídeo/química , Linhagem Celular , Células Clonais , Cruzamentos Genéticos , Epitopos/análise , Feminino , Citometria de Fluxo , Expressão Gênica , Complexo Principal de Histocompatibilidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Orthomyxoviridae/imunologia , Transfecção , Proteínas do Core Viral/biossíntese , Proteínas do Core Viral/química
8.
J Cell Biol ; 89(3): 585-92, 1981 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7251666

RESUMO

Locomoting polymorphonuclear leukocytes (PMNs) exhibit a morphological polarity. We demonstrate that they also exhibit a behavioral polarity in their responsiveness to chemotactic factor stimulation. This is demonstrated by (a) the pattern of their locomotion in a homogeneous concentration of chemotactic factors, (b) their responses to increases in the homogeneous concentration of chemotactic factors, and (c) their responses to changes in the direction of a chemotactic gradient. The behavioral polarity is not a function of the rate of locomotion of the particular stimulant used to orient the cells, but may reflect an asymmetric distribution of chemotactic receptors or the motile machinery. The polar behavior affects the chemotactic ability of PMNs. The data are discussed in relation to possible mechanisms of sensing a chemotactic gradient.


Assuntos
Quimiotaxia de Leucócito , Neutrófilos/fisiologia , Fatores Quimiotáticos/farmacologia , Humanos , Cinética , Neutrófilos/citologia , Oligopeptídeos/farmacologia , Pseudópodes/fisiologia
9.
Science ; 254(5032): 713-6, 1991 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-1948050

RESUMO

The generation of antigen-specific antitumor immunity is the ultimate goal in cancer immunotherapy. When cells from a spontaneously arising murine renal cell tumor were engineered to secrete large doses of interleukin-4 (IL-4) locally, they were rejected in a predominantly T cell-independent manner. However, animals that rejected the IL-4-transfected tumors developed T cell-dependent systemic immunity to the parental tumor. This systemic immunity was tumor-specific and primarily mediated by CD8+ T cells. Established parental tumors could be cured by the systemic immune response generated by injection of the genetically engineered tumors. These results provide a rationale for the use of lymphokine gene-transfected tumor cells as a modality for cancer therapy.


Assuntos
Carcinoma de Células Renais/terapia , Imunoterapia , Interleucina-4/genética , Neoplasias Renais/terapia , Engenharia de Proteínas , Animais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/patologia , Divisão Celular , Linhagem Celular , Interleucina-4/metabolismo , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Transplante de Neoplasias , Subpopulações de Linfócitos T/imunologia , Transfecção
10.
11.
Cancer Res ; 56(1): 21-6, 1996 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8548765

RESUMO

Presentation of antigenic peptides by MHC class II molecules to CD4+ T cells is critical to the generation of antitumor immunity. In an attempt to enhance MHC class II antigen processing, we linked the sorting signals of the lysosome-associated membrane protein (LAMP-1) to the cytoplasmic/nuclear human papilloma virus (HPV-16) E7 antigen, creating a chimera (Sig/E7/LAMP-1). Previously, we found that expression of this chimera in vitro and in vivo with a recombinant vaccinia vector targeted E7 to endosomal and lysosomal compartments and enhanced MHC class II presentation to CD4+ T cells compared to vaccinia expressing wild-type E7. In the current study, we tested these recombinant vaccinia for in vivo protection against an E7+ tumor, TC-1, which was derived from primary epithelial cells of C57BL/6 mice cotransformed with HPV-16 E6 and E7 and c-Ha-ras oncogenes. All mice vaccinated with 1 x 10(7) plaque-forming units of wild-type E7-vaccinia showed progressive tumor growth when challenged with a tumorigenic dose of TC-1 tumor cells; in contrast, 80% of mice vaccinated with the chimeric Sig/E7/LAMP1 vaccinia remained tumor free 3 months after tumor injection. Furthermore, treatment with the Sig/E7/LAMP-1 vaccinia vaccine cured mice with small established TC-1 tumors, whereas the wild-type E7-vaccinia showed no effect on this established tumor burden. These findings point out the therapeutic limitations of recombinant vaccinia expressing unmodified tumor antigens. Further, they demonstrate that modifications that reroute a cytosolic tumor antigen to the endosomal/lysosomal compartment can profoundly improve the in vivo therapeutic potency of recombinant vaccines.


Assuntos
Antígenos Virais de Tumores/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Neoplasias Experimentais/terapia , Vacinas Sintéticas , Animais , Apresentação de Antígeno , Sequência de Bases , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neoplasias Experimentais/imunologia , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
12.
Cancer Res ; 53(24): 5841-4, 1993 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8261390

RESUMO

Experimental studies using murine tumor models have demonstrated that potent systemic immunity can be generated using tumor vaccines engineered by gene transfer to secrete certain cytokines. The underlying physiological principle behind these strategies involves the sustained release of high doses of cytokine at the site of the tumor. In some cases, this paracrine approach appears to enhance tumor antigen presentation and avoids systemic cytokine toxicity. The widespread clinical use of autologous cytokine gene transduced tumor vaccines may be limited by the technical difficulty and labor intensity of individualized gene transfer. We have therefore explored an alternate approach to generating sustained release of cytokines local to the tumor cells. High doses of granulocyte-macrophage colony-stimulating factor encapsulated in cell-sized gelatin-chondroitin sulfate microspheres were mixed with irradiated tumor cells prior to s.c. injection. This vaccination scheme resulted in systemic anti-tumor immune responses comparable to granulocyte-macrophage colony-stimulating factor gene transduced tumor vaccines.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Imunoterapia Ativa , Melanoma Experimental/terapia , Animais , Biodegradação Ambiental , Preparações de Ação Retardada , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Interferon gama/administração & dosagem , Interferon gama/uso terapêutico , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Células Tumorais Cultivadas , Vacinação
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.
Cancer Res ; 59(20): 5160-8, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10537292

RESUMO

Vaccination with irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting gene-transduced cancer vaccines induces tumoricidal immune responses. In a Phase I human gene therapy trial, eight immunocompetent prostate cancer (PCA) patients were treated with autologous, GM-CSF-secreting, irradiated tumor vaccines prepared from ex vivo retroviral transduction of surgically harvested cells. Expansion of primary cultures of autologous vaccine cells was successful to meet trial specifications in 8 of 11 cases (73%); the yields of the primary culture cell limited the number of courses of vaccination. Side effects were pruritus, erythema, and swelling at vaccination sites. Vaccine site biopsies manifested infiltrates of dendritic cells and macrophages among prostate tumor vaccine cells. Vaccination activated new T-cell and B-cell immune responses against PCA antigens. T-cell responses, evaluated by assessing delayed-type hypersensitivity (DTH) reactions against untransduced autologous tumor cells, were evident in two of eight patients before vaccination and in seven of eight patients after treatment. Reactive DTH site biopsies manifested infiltrates of effector cells consisting of CD45RO+ T-cells, and degranulating eosinophils consistent with activation of both Th1 and Th2 T-cell responses. A distinctive eosinophilic vasculitis was evident near autologous tumor cells at vaccine sites, and at DTH sites. B-cell responses were also induced. Sera from three of eight vaccinated men contained new antibodies recognizing polypeptides of 26, 31, and 150 kDa in protein extracts from prostate cells. The 150-kDa polypeptide was expressed by LNCaP and PC-3 PCA cells, as well as by normal prostate epithelial cells, but not by prostate stromal cells. No antibodies against prostate-specific antigen were detected. These data suggest that both T-cell and B-cell immune responses to human PCA can be generated by treatment with irradiated, GM-CSF gene-transduced PCA vaccines.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Neoplasias da Próstata/terapia , Vacinas Sintéticas/imunologia , Linfócitos B/imunologia , Técnicas de Transferência de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Hipersensibilidade Tardia/etiologia , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Linfócitos T/imunologia , Vacinação
15.
Hum Gene Ther ; 10(12): 1983-91, 1999 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-10466632

RESUMO

Irradiated tumor cells transduced with the gene encoding the cytokine GM-CSF have been extensively studied as a vaccine formulation capable of priming systemic antitumor immune responses in the tumor-bearing host. In spite of the therapeutic promise of this vaccine strategy demonstrated in both animal models and early-phase clinical trials, clinical development has been limited by difficulties pertaining to the need to establish in culture the tumor of each patient and to perform individualized gene transfer. To circumvent these issues, we generated an HLA-negative human cell line producing large quantities of human GM-CSF for use as a universal bystander cell to be mixed with unmodified autologous tumor cells in the formulation of a vaccine. This line is easily propagated as a suspension culture in defined, serum-free medium. In a mouse model, we find that vaccination with a mixture of autologous tumor cells and an MHC-negative allogeneic GM-CSF-producing bystander cell primes antitumor immune responses that are equivalent or better than those achieved using autologous tumor cells directly transduced to secrete GM-CSF. This strategy greatly simplifies further clinical development of autologous tumor cell-based vaccines.


Assuntos
Vacinas Anticâncer , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Células Tumorais Cultivadas , Animais , Linhagem Celular , Transplante de Células , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Linfoma de Células B/terapia , Camundongos , Camundongos Endogâmicos BALB C , Transdução Genética
16.
Int J Oncol ; 12(1): 161-70, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9454900

RESUMO

Cancer cells genetically modified to secrete immunoregulatory cytokines offer great promise for human cancer treatment as tumor vaccines. However, in preclinical animal studies, large established cancer burdens have appeared difficult to eradicate with such vaccines. For example, lethally-irradiated GM-CSF-secreting CT26 colon carcinoma cell vaccine therapy tends to cure only animals bearing 1 x 10(5) wild-type CT26 cells or less. For many human cancers, antineoplastic chemotherapy can often significantly reduce systemic cancer burdens. Unfortunately, for most advanced metastatic solid organ cancers, such as cancers of the breast, colon, and prostate, antineoplastic drug treatments generally fail to effect cancer cures. Treatment regimens combining genetically-modified cancer cell vaccine therapy and antineoplastic chemotherapy have the potential to increase advanced cancer cure rates if antineoplastic drugs and drug combinations that do not inhibit vaccine-induced immune responses can be identified. To assess the potential immunomodulatory properties of commonly-used antineoplastic drugs that might be used in combination with cancer vaccine treatments, we studied the effects of the drugs on antitumor immune responses manifest by animals receiving lethally-irradiated GM-CSF-secreting CT26 cell vaccines. Immunomodulatory properties of the antineoplastic drugs were evaluated i) by monitoring drug effects on the generation of tumor-specific CD8+ cytotoxic T-lymphocytes (CTLs) in response to GM-CSF-secreting CT26 vaccine administration, ii) by determining drug effects on the resistance of vaccinated animals to subsequent challenge with lethal inoculac of CT26 cells, and iii) by evaluating combination drug and vaccine treatment efficacy against established CT26 tumors. Using this approach, doxorubicin was found to possess apparent immunostimulatory activities, depending on the dose and schedule of administration, while cyclophosphamide appeared immunosuppressive. The different immunomodulatory properties of doxorubicin and cyclophosphamide may be clinically relevant: combination doxorubicin and vaccine treatment of established CT26 cancers increased cure rates over that achieved with either agent alone, while combination cyclophosphamide and vaccine treatment of animals carrying CT26 tumors was no better in curing the animals than drug treatment alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Neoplasias do Colo/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias Experimentais/terapia , Adjuvantes Imunológicos/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Terapia Combinada , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Taxa de Sobrevida , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
17.
Cell Transplant ; 5(1): 77-91, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8665080

RESUMO

The ability to replace damaged myocardial tissue with new striated muscle would constitute a major advance in the treatment of diseases that irreversibly injure cardiac muscle cells. The creation of focal grafts of skeletal muscle has been reported following the intramural injection of skeletal myoblasts into both normal and injured myocardium. The goals of this study were to determine whether skeletal myoblast-derived cells can be engrafted into the murine heart following arterial delivery. The murine heart was seeded with genetically labeled C2C12 myoblasts introduced into the arterial circulation of the heart via a transventricular injection. A transventricular injection provided access to the coronary and systemic circulations. Implanted cells were characterized using histochemical staining for beta-galactosidase, immunofluorescent staining for muscle-specific antigens, and electron microscopy. Initially the injected cells were observed entrapped in myocardial capillaries. One week after injection myoblasts were present in the myocardial interstitium and were largely absent from the myocardial capillary bed. Implanted cells underwent myogenic development, characterized by the expression of a fast-twitch skeletal muscle sarcoendoplasmic reticulum calcium ATPase (SERCA1) and formation of myofilaments. Four months following injection myoblast-derived cells began to express a slow-twitch/cardiac protein, phospholamban, that is normally not expressed by C2C12 cells in vitro. Most surprisingly, regions of close apposition between LacZ labeled cells and native cardiomyocytes contained structures that resembled desmosomes, fascia adherens junctions, and gap junctions. The cardiac gap junction protein, connexin43, was localized to some of the interfaces between implanted cells and cardiomyocytes. Collectively, these findings suggest that arterially delivered myoblasts can be engrafted into the heart, and that prolonged residence in the myocardium may alter the phenotype of these skeletal muscle-derived cells. Further studies are necessary to determine whether arterial delivery of skeletal myoblasts can be developed as treatment for myocardial dysfunction.


Assuntos
Sobrevivência de Enxerto , Coração , Músculo Esquelético/transplante , Animais , Biomarcadores , Proteínas de Ligação ao Cálcio/análise , Proteínas de Ligação ao Cálcio/biossíntese , Linhagem Celular , Conexina 43/análise , Conexina 43/biossíntese , Escherichia coli , Masculino , Camundongos , Camundongos Endogâmicos C3H , Microscopia Eletrônica , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Proteínas Recombinantes/análise , Proteínas Recombinantes/biossíntese , Fatores de Tempo , Transfecção , Transplante Heterotópico/métodos , Transplante Heterotópico/fisiologia , beta-Galactosidase/análise , beta-Galactosidase/biossíntese
18.
Cancer Chemother Pharmacol ; 46 Suppl: S67-72, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10950151

RESUMO

When irradiated and administered intradermally as vaccines, cancer cells engineered to secrete high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) by gene transfer elicit potent anticancer immune responses in a variety of animal tumor models. Upon vaccination, antigens present in the cancer cells are phagocytosed and processed by skin dendritic cells. These dendritic cells then prime anticancer immune responses by presenting antigenic peptides to T cells. The immune responses generated are capable of eradicating small but lethal cancer cell inocula with minimal toxicity in preclinical animal tumor studies. To develop this vaccination strategy for the treatment of human genitourinary cancers, we have conducted phase I clinical trials using human genitourinary cancer cells as sources of cancer cell antigens. In the first human clinical trial of genetically engineered cancer cell vaccines, a phase I clinical trial of kidney cancer cell vaccines (n = 18), kidney cancer cells were removed at surgery, propagated briefly in vitro, and then genetically modified to secrete high levels of GM-CSF via ex vivo transduction with the retrovirus MFG-GM-CSF. After irradiation, the kidney cancer cells were administered as vaccines to 18 patients with advanced kidney cancers. Vaccine treatment, which caused few side effects, nonetheless appeared to trigger anticancer immune responses manifest as conversion of delayed-type hypersensitivity (DTH) skin responses against irradiated autologous cancer cells after vaccination. Biopsies of vaccine sites yielded findings reminiscent of biopsies from preclinical animal model studies, with evidence of vaccine cell recruitment of dendritic cells, T cells, and eosinophils. One patient with measurable kidney cancer metastases treated at the highest vaccine dose level experienced a partial treatment response. The bioactivity of GM-CSF-secreting autologous cancer cell vaccines was confirmed in a phase I clinical trial for prostate cancer (n = 8). Vaccine cells were prepared from surgically harvested prostate tumors by ex vivo transduction with MFG-GM-CSF in a manner similar to that used for the kidney cancer trial. Vaccine treatment was well tolerated and associated with induction of anticancer immunity as assessed using DTH skin testing. In addition, new antiprostate cancer cell antibodies were detected in serum samples from treated men as a consequence of vaccination. These first clinical trials of GM-CSF-secreting cancer cell vaccines for the treatment of genitourinary cancers have demonstrated both safety and bioactivity, in that very few side effects have been seen and anticancer immune responses have been detected. Future clinical studies will be required to assess vaccine treatment efficacy, refine vaccination dose and schedule, define the appropriate clinical context for the use of such vaccines, and ascertain optimal combinations involving vaccines and other local or systemic anticancer treatments.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Neoplasias Renais/imunologia , Neoplasias da Próstata/imunologia , Adulto , Idoso , Antígenos de Neoplasias/sangue , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Carcinoma de Células Renais/terapia , Relação Dose-Resposta Imunológica , Feminino , Técnicas de Transferência de Genes , Engenharia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Hipersensibilidade Tardia/imunologia , Neoplasias Renais/terapia , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/terapia , Retroviridae/genética
19.
Blood Cancer J ; 3: e145, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-24013666

RESUMO

The ability to target myeloid leukemia with immunotherapy would represent a significant therapeutic advance. We report here immunological analysis of clinical trials of primary and secondary vaccination with K562/GM-CSF immunotherapy in adult chronic phase chronic myeloid leukemia patients (CML-CP) with suboptimal responses to imatinib mesylate. Using serological analysis of recombinant cDNA expression libraries of K562 with autologous vaccinated patient serum, we have identified 12 novel chronic myeloid leukemia-associated antigens (LAAs). We show that clinical responses following K562/GM-CSF vaccination are associated with induction of high-titer antibody responses to multiple LAAs. We observe markedly discordant patterns of baseline and induced antibody responses in these identically vaccinated patients. No single antigen was recognized in all responses to vaccination. We demonstrate that an additional 'booster' vaccination series can be given safely to those with inadequate responses to initial vaccination, and is associated with more frequent induction of IgG responses to antigens overexpressed in K562 vaccine compared with primary CML-CP. Finally, those with induced immune responses to the same LAAs often shared HLA subtypes and patients with clinical responses following vaccination recognized a partially shared but non-identical spectrum of antigens; both findings have potentially significant implications for cancer vaccine immunotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA