Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Nat Commun ; 8(1): 592, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28928380

RESUMO

Immune checkpoint blockers (ICB) have become pivotal therapies in the clinical armamentarium against metastatic melanoma (MMel). Given the frequency of immune related adverse events and increasing use of ICB, predictors of response to CTLA-4 and/or PD-1 blockade represent unmet clinical needs. Using a systems biology-based approach to an assessment of 779 paired blood and tumor markers in 37 stage III MMel patients, we analyzed association between blood immune parameters and the functional immune reactivity of tumor-infiltrating cells after ex vivo exposure to ICB. Based on this assay, we retrospectively observed, in eight cohorts enrolling 190 MMel patients treated with ipilimumab, that PD-L1 expression on peripheral T cells was prognostic on overall and progression-free survival. Moreover, detectable CD137 on circulating CD8+ T cells was associated with the disease-free status of resected stage III MMel patients after adjuvant ipilimumab + nivolumab (but not nivolumab alone). These biomarkers should be validated in prospective trials in MMel.The clinical management of metastatic melanoma requires predictors of the response to checkpoint blockade. Here, the authors use immunological assays to identify potential prognostic/predictive biomarkers in circulating blood cells and in tumor-infiltrating lymphocytes from patients with resected stage III melanoma.

2.
Cell Death Dis ; 7: e2302, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27441655

RESUMO

Inflammatory bowel disease (IBD) is an immunoregulatory disorder, associated with a chronic and inappropriate mucosal immune response to commensal bacteria, underlying disease states such as ulcerative colitis (UC) and Crohn's disease (CD) in humans. Granzyme M (GrzM) is a serine protease expressed by cytotoxic lymphocytes, in particular natural killer (NK) cells. Granzymes are thought to be involved in triggering cell death in eukaryotic target cells; however, some evidence supports their role in inflammation. The role of GrzM in the innate immune response to mucosal inflammation has never been examined. Here, we discover that patients with UC, unlike patients with CD, display high levels of GrzM mRNA expression in the inflamed colon. By taking advantage of well-established models of experimental UC, we revealed that GrzM-deficient mice have greater levels of inflammatory indicators during dextran sulfate sodium (DSS)-induced IBD, including increased weight loss, greater colon length reduction and more severe intestinal histopathology. The absence of GrzM expression also had effects on gut permeability, tissue cytokine/chemokine dynamics, and neutrophil infiltration during disease. These findings demonstrate, for the first time, that GrzM has a critical role during early stages of inflammation in UC, and that in its absence colonic inflammation is enhanced.


Assuntos
Colite Ulcerativa/imunologia , Colite/imunologia , Doença de Crohn/imunologia , Granzimas/imunologia , Imunidade Inata , Animais , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Colo/imunologia , Colo/patologia , Doença de Crohn/genética , Doença de Crohn/patologia , Sulfato de Dextrana , Feminino , Expressão Gênica , Granzimas/deficiência , Granzimas/genética , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Permeabilidade , RNA Mensageiro/genética , RNA Mensageiro/imunologia
3.
Immunol Cell Biol ; 93(2): 198-212, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385067

RESUMO

Interleukin-17A (IL-17A) is a pro-inflammatory cytokine that has an important role at mucosal sites in a wide range of immune responses including infection, allergy and auto-immunity. γδ T cells are recognized as IL-17 producers, but based on the level of CD3 expression, we now define the remarkable ability of a CD3(bright) γδ T-cell subset with an effector memory phenotype to rapidly produce IL-17A, but not interferon-γ. CD3(bright) γδ T cells uniformly express the canonical germline encoded Vγ6/Vδ1(+) T-cell receptor. They are widely distributed with a preferential representation in the lungs and skin are negatively impacted in the absence of retinoic acid receptor-related orphan receptor gammat expression or endogenous flora. This population responded rapidly to various stimuli in a mechanism involving IL-23 and NOD-like receptor family, pyrin domain containing 3 (NLRP3)-inflammasome-dependent IL-1ß. Finally, we demonstrated that IL-17-producing CD3(bright) γδ T cells responded promptly and strongly to pneumococcal infection and during skin inflammation. Here, we propose a new way to specifically analyze IL-17-producing Vγ6/Vδ1(+) T cells based on the level of CD3 signals. Using this gating strategy, our data reinforce the crucial role of this γδ T-cell subset in respiratory and skin disorders.


Assuntos
Complexo CD3/metabolismo , Interleucina-17/biossíntese , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/imunologia , Sequência de Aminoácidos , Aminoquinolinas/farmacologia , Animais , Complexo CD3/química , Proteínas de Transporte/metabolismo , Células Germinativas/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Imiquimode , Imunidade , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Interleucina-23 , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteína 3 que Contém Domínio de Pirina da Família NLR , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fenótipo , Pele/efeitos dos fármacos , Pele/imunologia , Linfócitos T/efeitos dos fármacos
4.
Ann Oncol ; 25(8): 1544-50, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24608200

RESUMO

BACKGROUND: We have previously shown the prognostic importance of tumor-infiltrating lymphocytes (TILs) in newly diagnosed triple-negative breast cancer (TNBC) using tumor samples from a large clinical trial cohort. In this study, we aimed to validate these findings and also investigate associations with trastuzumab benefit in HER2-overexpressing disease (HER2+). PATIENTS AND METHODS: A prospective-retrospective study was conducted using samples from the FinHER adjuvant, phase III trial that enrolled 1010 early-stage BC patients, 778 of whom were HER2-nonamplified. Those with HER2+ disease (n = 232) were randomized to 9 weeks of trastuzumab or no trastuzumab in addition to chemotherapy. Two pathologists independently quantified stromal TILs in 935 (92.6%) available slides. The primary end point of distant disease-free survival (DDFS) and interactions with trastuzumab were studied in Cox regression models. RESULTS: Confirming our previous findings, in TNBC (n = 134) each 10% increase in TILs was significantly associated with decreased distant recurrence in TNBC; for DDFS the hazard ratio adjusted for clinicopathological factors: 0.77; 95% confidence interval (CI) 0.61-0.98, P = 0.02. In HER2+ BC (n = 209), each 10% increase in lymphocytic infiltration was significantly associated with decreased distant recurrence in patients randomized to the trastuzumab arm (DDFS P interaction = 0.025). CONCLUSIONS: Higher levels of TILs present at diagnosis were significantly associated with decreased distant recurrence rates in primary TNBC. These results confirm our previous data and further support that TILs should be considered as a robust prognostic factor in this BC subtype. We also report for the first time an association between higher levels of TILs and increased trastuzumab benefit in HER2+ disease. Further research into why some TN and HER2+ BCs can or cannot generate a host antitumor immune response and how trastuzumab can favorably alter the immune microenvironment is warranted.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Farmacológicos , Biomarcadores Tumorais , Linfócitos do Interstício Tumoral/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/imunologia , Adulto , Idoso , Biomarcadores Farmacológicos/análise , Biomarcadores Tumorais/análise , Quimioterapia Adjuvante , Feminino , Finlândia , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Prognóstico , Trastuzumab , Resultado do Tratamento , Neoplasias de Mama Triplo Negativas/diagnóstico , Neoplasias de Mama Triplo Negativas/patologia
5.
Cell Death Dis ; 5: e1115, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24625974

RESUMO

Granzymes are generally recognized for their capacity to induce various pathways of perforin-dependent target cell death. Within this serine protease family, Granzyme M (GrzM) is unique owing to its preferential expression in innate effectors such as natural killer (NK) cells. During Listeria monocytogenes infection, we observed markedly reduced secretion of macrophage inflammatory protein-1 alpha (MIP-1α) in livers of GrzM-deficient mice, which resulted in significantly impaired NK cell recruitment. Direct stimulation with IL-12 and IL-15 demonstrated that GrzM was required for maximal secretion of active MIP-1α. This effect was not due to reduced protein induction but resulted from heightened intracellular accumulation of MIP-1α, with reduced release. These results demonstrate that GrzM is a critical mediator of innate immunity that can regulate chemotactic networks and has an important role in the initiation of immune responses and pathogen control.


Assuntos
Quimiocina CCL3/metabolismo , Granzimas/metabolismo , Imunidade Inata , Células Matadoras Naturais/enzimologia , Listeriose/enzimologia , Animais , Células Cultivadas , Quimiotaxia de Leucócito , Técnicas de Cocultura , Modelos Animais de Doenças , Granzimas/deficiência , Granzimas/genética , Humanos , Interleucinas/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/microbiologia , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Listeriose/genética , Listeriose/imunologia , Listeriose/microbiologia , Camundongos , Camundongos Knockout , Fatores de Tempo
6.
Cell Death Differ ; 21(6): 876-87, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24488096

RESUMO

Cytotoxic lymphocytes (CLs) contain lysosome-related organelles (LROs) that perform the normal degradative functions of the lysosome, in addition to storage and release of powerful cytotoxins employed to kill virally infected or abnormal cells. Among these cytotoxins is granzyme B (GrB), a protease that has also been implicated in activation (restimulation)-induced cell death of natural killer (NK) and T cells, but the underlying mechanism and its regulation are unclear. Here we show that restimulation of previously activated human or mouse lymphocytes induces lysosomal membrane permeabilisation (LMP), followed by GrB release from LROs into the CL cytosol. The model lysosomal stressors sphingosine and Leu-Leu-methyl-ester, and CLs from gene-targeted mice were used to show that LMP releases GrB in both a time- and concentration-dependent manner, and that the liberated GrB is responsible for cell death. The endogenous GrB inhibitor Serpinb9 (Sb9) protects CLs against LMP-induced death but is decreasingly effective as the extent of LMP increases. We also used these model stressors to show that GrB is the major effector of LMP-mediated death in T cells, but that in NK cells additional effectors are released, making GrB redundant. We found that limited LMP and GrB release occurs constitutively in proliferating lymphocytes and in NK cells engaged with targets in vitro. In Ectromelia virus-infected lymph nodes, working NK cells lacking Sb9 are more susceptible to GrB-mediated death. Taken together, these data show that a basal level of LMP occurs in proliferating and activated lymphocytes, and is increased on restimulation. LMP releases GrB from LROs into the lymphocyte cytoplasm and its ensuing interaction with Sb9 dictates whether or not the cell survives. The GrB-Sb9 nexus may therefore represent an additional mechanism of limiting lymphocyte lifespan and populations.


Assuntos
Morte Celular/efeitos dos fármacos , Granzimas/metabolismo , Serpinas/metabolismo , Estresse Fisiológico/genética , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/patologia , Camundongos , Esfingosina/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
7.
Cell Death Differ ; 21(1): 5-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23579243

RESUMO

Protection against cellular stress from various sources, such as nutritional, physical, pathogenic, or oncogenic, results in the induction of both intrinsic and extrinsic cellular protection mechanisms that collectively limit the damage these insults inflict on the host. The major extrinsic protection mechanism against cellular stress is the immune system. Indeed, it has been well described that cells that are stressed due to association with viral infection or early malignant transformation can be directly sensed by the immune system, particularly natural killer (NK) cells. Although the ability of NK cells to directly recognize and respond to stressed cells is well appreciated, the mechanisms and the breadth of cell-intrinsic responses that are intimately linked with their activation are only beginning to be uncovered. This review will provide a brief introduction to NK cells and the relevant receptors and ligands involved in direct responses to cellular stress. This will be followed by an in-depth discussion surrounding the various intrinsic responses to stress that can naturally engage NK cells, and how therapeutic agents may induce specific activation of NK cells and other innate immune cells by activating cellular responses to stress.


Assuntos
Células Matadoras Naturais/metabolismo , Estresse Fisiológico/imunologia , Proteínas de Choque Térmico/metabolismo , Humanos , Sistema Imunitário/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Ligantes , MicroRNAs/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Oncogenes , Receptores de Células Matadoras Naturais/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Viroses/imunologia , Viroses/metabolismo , Viroses/patologia
8.
Cell Death Differ ; 21(1): 50-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23744294

RESUMO

Immunogenic cell death induced by cytotoxic compounds contributes to the success of selected chemotherapies by eliciting a protective anticancer immune response, which is mediated by CD4(+) and CD8(+) T cells producing interferon-γ. In many instances, cancer progression is associated with high titers of tumor-specific antibodies, which become detectable in the serum, but whose functional relevance is elusive. Here, we explored the role of humoral immune responses in the anticancer efficacy of anthracyclines. Chemotherapy reduced the number of tumor-infiltrating B cells, and failed to promote humoral responses against immunodominant tumor antigens. Although anthracycline-based anticancer chemotherapies failed in T cell-deficient mice, they successfully reduced the growth of cancers developing in mice lacking B lymphocytes (due to the injection of a B-cell-depleting anti-CD20 antibody), immunoglobulins (Igs) or Ig receptors (Fc receptor) due to genetic manipulations. These results suggest that the humoral arm of antitumor immunity is dispensable for the immune-dependent therapeutic effect of anthracyclines against mouse sarcoma. In addition, we show here that the titers of IgA and IgG antibodies directed against an autoantigen appearing at the cell surface of tumor cells post chemotherapy (calreticulin, CRT) did not significantly increase in patients treated with anthracyclines, and that anti-CRT antibodies had no prognostic or predictive significance. Collectively, our data indicate that humoral anticancer immune responses differ from cellular responses in, thus far, that they do not contribute to the success of anthracycline-mediated anticancer therapies in human breast cancers and mouse sarcomas.


Assuntos
Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Imunidade Humoral , Animais , Antraciclinas/toxicidade , Anticorpos/imunologia , Anticorpos/farmacologia , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Calreticulina/imunologia , Calreticulina/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Receptores Fc/metabolismo , Sarcoma/tratamento farmacológico , Linfócitos T Citotóxicos/imunologia
9.
Oncogene ; 29(39): 5346-58, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20661219

RESUMO

Adenosine triphosphate (ATP) is actively released in the extracellular environment in response to tissue damage and cellular stress. Through the activation of P2X and P2Y receptors, extracellular ATP enhances tissue repair, promotes the recruitment of immune phagocytes and dendritic cells, and acts as a co-activator of NLR family, pyrin domain-containing 3 (NLRP3) inflammasomes. The conversion of extracellular ATP to adenosine, in contrast, essentially through the enzymatic activity of the ecto-nucleotidases CD39 and CD73, acts as a negative-feedback mechanism to prevent excessive immune responses. Here we review the effects of extracellular ATP and adenosine on tumorigenesis. First, we summarize the functions of extracellular ATP and adenosine in the context of tumor immunity. Second, we present an overview of the immunosuppressive and pro-angiogenic effects of extracellular adenosine. Third, we present experimental evidence that extracellular ATP and adenosine receptors are expressed by tumor cells and enhance tumor growth. Finally, we discuss recent studies, including our own work, which suggest that therapeutic approaches that promote ATP-mediated activation of inflammasomes, or inhibit the accumulation of tumor-derived extracellular adenosine, may constitute effective new means to induce anticancer activity.


Assuntos
Trifosfato de Adenosina/fisiologia , Adenosina/fisiologia , Neoplasias/metabolismo , Trifosfato de Adenosina/farmacologia , Espaço Extracelular/metabolismo , Humanos , Imunidade , Terapia de Imunossupressão , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais
10.
Gene Ther ; 17(9): 1105-16, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20428216

RESUMO

The anti-tumor efficacy of adoptively transferred T cells requires their in vivo persistence and memory polarization. It is unknown if human chimeric antigen receptor (CAR)-expressing T cells can also undergo memory polarization. We examined the functional status of CAR CD8(+) T cells, re-directed to Lewis Y antigen (LeY-T), throughout a period of ex vivo expansion. Immediately before culture CD8(+) T cells comprised a mixture of phenotypes including naive (CD45RA(+)/CCR7(+)/CD27(+)/CD28(+)/perforin-), central memory (CM, CD45RA(-)/CCR7(lo)/CD27(+)/CD28(+)/perforin(lo)), effector memory (EM, CD45RA(-)/CCR7(-)/CD27(+)/CD28(+)/perforin(mod)) and effector (Eff, CD45RA(+)/CCR7(-)/CD27(-)/CD28(-)/perforin(hi)) cells. After transduction and expansion culture of peripheral blood mononuclear cells from normal donors or multiple myeloma patients, CD8(+) LeY-T cells polarized to EM- and CM-like phenotype. CD8(+) LeY-T cells differed from starting CD8(+) CM and EM T cells in that CD27, but not CD28, was downregulated. In addition, CD8(+) LeY-T cells expressed high levels of perforin, similar to starting CD8(+) Eff. CD8(+) LeY-T cells also showed hallmarks of both memory and Eff function, underwent homeostatic proliferation in response to interleukin (IL)-15, and showed interferon (IFN)-γ production and cytotoxicity in response to Le-Y antigen on OVCAR-3 (human ovarian adenocarcinoma) cells. This study confirms CD8(+) LeY-T cells have a CM- and EM-like phenotype and heterogeneous function consistent with potential to persist in vivo after adoptive transfer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Memória Imunológica , Receptores de Antígenos/genética , Antígenos CD28/imunologia , Proliferação de Células , Humanos , Interferon gama/metabolismo , Antígenos Comuns de Leucócito/imunologia , Fenótipo , Receptores de Antígenos/imunologia , Receptores de Antígenos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
11.
Gene Ther ; 17(5): 678-86, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20200563

RESUMO

We have evaluated the carbohydrate antigen Lewis(Y) (Le(Y)) as a potential target for T-cell immunotherapy of hematological neoplasias. Analysis of 81 primary bone marrow samples revealed moderate Le(Y) expression on plasma cells of myeloma patients and myeloblasts of patients with acute myeloid leukemia (AML) (52 and 46% of cases, respectively). We developed a retroviral vector construct encoding a chimeric T-cell receptor that recognizes the Le(Y) antigen in a major histocompatibility complex-independent manner and delivers co-stimulatory signals to achieve T-cell activation. We have shown efficient transduction of peripheral blood-derived T cells with this construct, resulting in antigen-restricted interferon-gamma secretion and cell lysis of Le(Y)-expressing tumor cells. In vivo activity of gene-modified T cells was demonstrated in the delayed growth of myeloma xenografts in NOD/SCID mice, which prolonged survival. Therefore, targeting Le(Y)-positive malignant cells with T cells expressing a chimeric receptor recognizing Le(Y) was effective both in vitro and in a myeloma mouse model. Consequently, we plan to use T cells manufactured under Good Manufacturing Practice conditions in a phase I immunotherapy study for patients with Le(Y)-positive myeloma or AML.


Assuntos
Terapia Genética/métodos , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/terapia , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Mieloma Múltiplo/terapia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/transplante , Animais , Feminino , Vetores Genéticos , Humanos , Interferon gama/metabolismo , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Antígenos de Linfócitos T/imunologia , Retroviridae/genética , Linfócitos T/imunologia , Transdução Genética
12.
Leukemia ; 24(1): 22-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19907437

RESUMO

Immunomodulatory drugs (IMiDs) are thalidomide analogues, which possess pleiotropic anti-myeloma properties including immune-modulation, anti-angiogenic, anti-inflammatory and anti-proliferative effects. Their development was facilitated by an improved understanding in myeloma (MM) biology and initiated a profound shift in the therapeutic approach towards MM. Despite the diverse effects of IMiDs in vitro, the relative contribution of each effect towards their ultimate anti-MM activity is still unclear. Based on in vitro data, it appears that anti-proliferative effects and downregulation of crucial cytokines are their most important anti-MM attributes. Although the co-stimulatory effects on T and NK cells have been heralded as a unique and important property of IMiDs towards enhancing anti-MM immune activity, these in vitro effects have yet to be firmly corroborated in vivo. Much is yet to be elucidated regarding the complex interplay of immunomodulatory cytokines that occurs in vivo, which ultimately dictates the net effects of IMiDs in MM-the understanding of which is necessary to facilitate optimal manipulation of these drugs in future MM management.


Assuntos
Fatores Imunológicos/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Talidomida/análogos & derivados , Inibidores da Angiogênese/farmacologia , Anti-Inflamatórios/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Antineoplásicos/farmacologia , Fatores de Transcrição Forkhead/análise , Humanos , Fatores Imunológicos/uso terapêutico , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Mieloma Múltiplo/imunologia , Osteoclastos/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
13.
Gene Ther ; 15(14): 1056-66, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18369322

RESUMO

There is considerable concern regarding the transforming potential of retroviral vectors currently used for gene therapy, with evidence that retroviral integration can lead to leukemia in recipients of gene-modified stem cells. However, it is not clear whether retroviral-mediated transduction of T cells can lead to malignancy. We transduced mouse T cells with a Moloney murine retroviral gene construct and transferred them into congenic mice, which were preconditioned to enhance the engraftment of transferred T cells. Recipients were then observed long-term for evidence of cancer. Transferred T cells persisted in mice throughout life at levels up to 17% with gene copy numbers up to 5.89 x 10(5) per million splenocytes. Mice receiving gene-modified T cells developed tumors at a similar rate as control mice that did not receive T cells, and tumors in both groups of mice were of a similar range of histologies. Hematological malignancies comprised approximately 60% of cancers, and the remaining cancers consisted largely of carcinomas. Importantly, the incidence of lymphomas was similar in both groups of mice, and no lymphomas were found to be of donor T-cell origin. This study indicates that the use of retroviral vectors to transduce T cells does not lead to malignant transformation.


Assuntos
Transferência Adotiva , Terapia Genética/efeitos adversos , Vetores Genéticos/administração & dosagem , Vírus da Leucemia Murina de Moloney/fisiologia , Linfócitos T/virologia , Animais , Transformação Celular Viral , Leucemia/virologia , Linfoma/virologia , Camundongos , Camundongos SCID , Vírus da Leucemia Murina de Moloney/genética , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T/transplante , Tempo , Transdução Genética/métodos , Transgenes
14.
Curr Top Microbiol Immunol ; 314: 293-323, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17593666

RESUMO

CD1d-restricted T cells (NKT cells) are potent regulators of a broad range of immune responses. In particular, an abundance of research has focussed on the role of NKT cells in tumor immunity. This field of research has been greatly facilitated by the finding of agonist ligands capable of potently stimulating NKT cells and also animal models where NKT cells have been shown to play a natural role in the surveillance of tumors. Herein, we review the capability of NKT cells to promote the rejection of tumors and the mechanisms by which this occurs. We also highlight a growing field of research that has found that NKT cells are capable of suppressing anti-tumor immunity and discuss the progress to date for the immunotherapeutic use of NKT cells.


Assuntos
Antígenos CD1/metabolismo , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Animais , Ensaios Clínicos Fase I como Assunto , Modelos Animais de Doenças , Galactosilceramidas/imunologia , Galactosilceramidas/uso terapêutico , Humanos , Células Matadoras Naturais/metabolismo , Camundongos , Monitorização Imunológica , Neoplasias/terapia
15.
Oncogene ; 26(25): 3745-57, 2007 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-17530027

RESUMO

Deregulated cell death pathways may lead to the development of cancer, and induction of tumor cell apoptosis is the basis of many cancer therapies. Knowledge accumulated concerning the molecular mechanisms of apoptotic cell death has aided the development of new therapeutic strategies to treat cancer. Signals through death receptors of the tumor necrosis factor (TNF) superfamily have been well elucidated, and death receptors are now one of the most attractive therapeutic targets in cancer. In particular, DR5 and DR4, death receptors of TNF-related apoptosis-inducing ligand (TRAIL or Apo2L), are interesting targets of antibody-based therapy, since TRAIL may also bind decoy receptors that may prevent TRAIL-mediated apoptosis, whereas TRAIL ligand itself selectively induces apoptosis in cancer cells. Here, we review the potential therapeutic utility of agonistic antibodies against DR5 and DR4 and discuss the possible extension of this single-antibody-based strategy when combined with additional modalities that either synergizes to cause enhanced apoptosis or further engage the cellular immune response. Rational design of antibody-based therapies combining the induction of tumor cell apoptosis and activation of tumor-specific adaptive immunity enables promotion of distinct steps of the antitumor immune response, thereby enhancing tumor-specific lymphocytes that can eradicate TRAIL/DR5-resistant mutating, large established and heterogeneous tumors in a manner that does not require the definition of individual tumor-specific antigens.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores de Morte Celular/metabolismo , Animais , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/uso terapêutico , Progressão da Doença , Humanos , Imunoterapia , Neoplasias/imunologia , Neoplasias/patologia , Receptores de Morte Celular/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
16.
Proc Natl Acad Sci U S A ; 104(19): 8071-6, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17470784

RESUMO

Histone deacetylase inhibitors (HDACi) can elicit a range of biological responses that affect tumor growth and survival, including inhibition of cell cycle progression, induction of tumor cell-selective apoptosis, suppression of angiogenesis, and modulation of immune responses, and show promising activity against hematological malignancies in clinical trials. Using the Emu-myc model of B cell lymphoma, we screened tumors with defined genetic alterations in apoptotic pathways for therapeutic responsiveness to the HDACi vorinostat. We demonstrated a direct correlation between induction of tumor cell apoptosis in vivo and therapeutic efficacy. Vorinostat did not require p53 activity or a functional death receptor pathway to kill Emu-myc lymphomas and mediate a therapeutic response but depended on activation of the intrinsic apoptotic pathway with the proapoptotic BH3-only proteins Bid and Bim playing an important role. Our studies provide important information regarding the mechanisms of action of HDACi that have broad implications regarding stratification of patients receiving HDACi therapy alone or in combination with other anticancer agents.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/uso terapêutico , Linfoma de Células B/tratamento farmacológico , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/fisiologia , Proteína 11 Semelhante a Bcl-2 , Genes myc , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Vorinostat , Proteína bcl-X/fisiologia
17.
Cancer Gene Ther ; 14(5): 499-508, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17332777

RESUMO

Activation and expansion of T cells are important in disease resolution, but tumors do not usually satisfy these immune requirements. Therefore, we employed a novel strategy whereby dual-specific T cells were generated that could respond to both tumor and influenza virus, reasoning that immunization with influenza virus would activate and expand tumor-specific cells, and inhibit tumor growth. Dual-specific T cells were generated by gene modification of influenza virus-specific mouse T cells with a chimeric gene-encoding reactivity against the erbB2 tumor-associated antigen. Dual-specific T cells were demonstrated to respond against both tumor and influenza in vitro, and expanded in vitro in response to influenza to a much greater degree than in response to tumor cells. Following adoptive transfer and immunization of tumor-bearing mice with influenza virus, dual-specific T cells expanded greatly in numbers in the peritoneal cavity and spleen. This resulted in a significant increase in time of survival of mice. However, tumors were not eradicated, which may have been due to the observed poor penetration of tumor by T cells. This is the first demonstration that the potent immunogenic nature of an infectious agent can be utilized to directly impact on T-cell expansion and activity against tumor in vivo.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias Mamárias Animais/terapia , Orthomyxoviridae/imunologia , Receptor ErbB-2/antagonistas & inibidores , Linfócitos T/transplante , Animais , Antígenos de Neoplasias/imunologia , Camundongos , Camundongos Endogâmicos , Receptor ErbB-2/imunologia , Linfócitos T/imunologia
18.
Cell Death Differ ; 11(9): 1028-37, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15131592

RESUMO

P-glycoprotein (P-gp) can induce multidrug resistance (MDR) through the ATP-dependent efflux of chemotherapeutic agents. We have previously shown that P-gp can inhibit nondrug apoptotic stimuli by suppressing the activation of caspases. To determine if this additional activity is functionally linked to ATP hydrolysis, we expressed wild-type and ATPase-mutant P-gp and showed that cells expressing mutant P-gp could not efflux chemotherapeutic drugs but remained relatively resistant to apoptosis. CEM lymphoma cells expressing mutant P-gp treated with vincristine showed a decrease in the fraction of cells with apoptotic morphology, cytochrome c release from the mitochondria and suppression of caspase activation, yet still accumulated in mitosis and showed a loss of clonogenic potential. The loss of clonogenicity in vincristine-treated cells expressing mutant P-gp was associated with accumulation of cells in mitosis and the presence of multinucleated cells consistent with mitotic catastrophe. The antiapoptotic effect of mutant P-gp was not affected by antibodies that inhibit the efflux function of the protein. These data are consistent with a dual activity model for P-gp-induced MDR involving both ATPase-dependent drug efflux and ATPase-independent inhibition of apoptosis. The structure-function analyses described herein provide novel insight into the mechanisms of action of P-gp in mediating MDR.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Trifosfato de Adenosina/metabolismo , Caspases/metabolismo , Adenosina Trifosfatases/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Sobrevivência Celular , Citocromos c/metabolismo , Análise Mutacional de DNA , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hidrólise , Ácidos Hidroxâmicos/farmacologia , Idarubicina/farmacologia , Linfoma/tratamento farmacológico , Mitose , Mutação , Retroviridae/genética , Relação Estrutura-Atividade , Fatores de Tempo , Vincristina/farmacologia
19.
Cell Death Differ ; 9(11): 1266-72, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12404126

RESUMO

Previous studies by our laboratory have shown that the drug transporter protein P-glycoprotein, P-gp, can specifically inhibit Fas-induced caspase-3 activation and apoptosis. Importantly, inhibition of both caspase-3 activation and cell death could be reversed by pharmacological and antibody inhibitors of P-gp function. However, the molecular mechanisms underpinning P-gp-mediated resistance to Fas-induced cell death and caspase activation remained unknown. We therefore sought to identify the point(s) within the death receptor pathway at which P-gp exerted its inhibitory effect and to determine whether the ATPase activity of P-gp was required. Structure-function analysis determined that ATP hydrolysis was necessary for P-gp to confer resistance to Fas-induced caspase activation and cell death. Importantly, although both FADD and caspase-8 were recruited to the Death Inducing Signal Complex (DISC) in wild-type P-gp expressing cells following Fas ligation, subsequent activation of caspase-8 at the DISC was inhibited. The ability of P-gp to inhibit caspase-8 activation was also ATP dependent. These studies demonstrate that P-gp inhibits Fas-induced caspase-8 activation but not formation of the DISC and that this activity of P-gp is dependent on ATP hydrolysis.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Caspases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor fas/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Caspase 8 , Caspase 9 , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Proteína de Domínio de Morte Associada a Fas , Vetores Genéticos , Humanos , Retroviridae , Transdução Genética
20.
Proc Natl Acad Sci U S A ; 98(19): 10833-8, 2001 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-11535817

RESUMO

Many chemotherapeutic agents induce mitochondrial-membrane disruption to initiate apoptosis. However, the upstream events leading to drug-induced mitochondrial perturbation have remained poorly defined. We have used a variety of physiological and pharmacological inhibitors of distinct apoptotic pathways to analyze the manner by which suberoylanilide hydroxamic acid (SAHA), a chemotherapeutic agent and histone deacetylase inhibitor, induces cell death. We demonstrate that SAHA initiates cell death by inducing mitochondria-mediated death pathways characterized by cytochrome c release and the production of reactive oxygen species, and does not require the activation of key caspases such as caspase-8 or -3. We provide evidence that mitochondrial disruption is achieved by means of the cleavage of the BH3-only proapoptotic Bcl-2 family member Bid. SAHA-induced Bid cleavage was not blocked by caspase inhibitors or the overexpression of Bcl-2 but did require the transcriptional regulatory activity of SAHA. These data provide evidence of a mechanism of cell death mediated by transcriptional events that result in the cleavage of Bid, disruption of the mitochondrial membrane, and production of reactive oxygen species to induce cell death.


Assuntos
Antineoplásicos/metabolismo , Apoptose , Proteínas de Transporte/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/farmacologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Proteínas de Transporte/genética , Caspase 10 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transcrição Gênica , Células Tumorais Cultivadas , Vorinostat
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...