Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Int J Cancer ; 128(11): 2735-47, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20715169

RESUMO

The recent development of novel targeted anticancer therapeutics such as histone deacetylase inhibitors (HDACi) and activators of the TRAIL pathway provide opportunities for the introduction of new treatment regimens in oncology. HDACi and recombinant TRAIL or agonistic anti-TRAIL receptor antibodies have been shown to induce synergistic tumor cell apoptosis and some therapeutic activity in vivo. Herein, we have used syngeneic preclinical models of human solid cancers to demonstrate that the HDACi panobinostat can sensitize tumor cells to apoptosis mediated by the anti-mouse TRAIL receptor antibody MD5-1. We demonstrate that the combination of panobinostat and MD5-1 can eradicate tumors grown subcutaneously and orthotopically in immunocompetent mice, while single agent treatment has minimal effect. However, escalation of the dose of panobinostat to enhance antitumor activity resulted in on-target MD5-1-mediated gastrointestinal toxicities that were fatal to the treated mice. Studies performed in mice with knockout of the TRAIL receptor showed that these mice could tolerate doses of the panobinostat/MD5-1 combination that were lethal in wild type mice resulting in superior tumor clearance. Given that clinical studies using HDACi and activators of the TRAIL pathway have been initiated, our preclinical data highlight the potential toxicities that could limit the use of such a treatment regimen. Our studies also demonstrate the power of using syngeneic in vivo tumor models as physiologically relevant preclinical systems to test the antitumor effects and identify potential side effects of novel anticancer regimens.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Neoplasias Mamárias Experimentais/terapia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Animais , Apoptose , Western Blotting , Terapia Combinada , Sinergismo Farmacológico , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Indóis , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Panobinostat , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Proc Natl Acad Sci U S A ; 105(32): 11317-22, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18685088

RESUMO

Histone deacetylase inhibitors (HDACi) and agents such as recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and agonistic anti-TRAIL receptor (TRAIL-R) antibodies are anticancer agents that have shown promise in preclinical settings and in early phase clinical trials as monotherapies. Although HDACi and activators of the TRAIL pathway have different molecular targets and mechanisms of action, they share the ability to induce tumor cell-selective apoptosis. The ability of HDACi to induce expression of TRAIL-R death receptors 4 and 5 (DR4/DR5), and induce tumor cell death via the intrinsic apoptotic pathway provides a molecular rationale to combine these agents with activators of the TRAIL pathway that activate the alternative (death receptor) apoptotic pathway. Herein, we demonstrate that the HDACi vorinostat synergizes with the mouse DR5-specific monoclonal antibody MD5-1 to induce rapid and robust tumor cell apoptosis in vitro and in vivo. Importantly, using a preclinical mouse breast cancer model, we show that the combination of vorinostat and MD5-1 is safe and induces regression of established tumors, whereas single agent treatment had little or no effect. Functional analyses revealed that rather than mediating enhanced tumor cell apoptosis via the simultaneous activation of the intrinsic and extrinsic apoptotic pathways, vorinostat augmented MD5-1-induced apoptosis concomitant with down-regulation of the intracellular apoptosis inhibitor cellular-FLIP (c-FLIP). These data demonstrate that combination therapies involving HDACi and activators of the TRAIL pathway can be efficacious for the treatment of cancer in experimental mouse models.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases , Neoplasias Mamárias Experimentais/tratamento farmacológico , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Animais , Anticorpos Monoclonais/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/biossíntese , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Proteínas de Neoplasias/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Vorinostat
3.
Cancer Lett ; 280(2): 125-33, 2009 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-19359091

RESUMO

Histone deacetylase inhibitors (HDACi) are anti-cancer drugs that have moved rapidly through clinical development and in 2006 vorinostat (SAHA, Zolinza) was given FDA approval for the treatment of cutaneous T cell lymphoma. Class I, II and IV HDACs that are targets for these compounds deacetylate histone proteins, resulting in chromatin remodelling and altered gene transcription. In addition, numerous non-histone proteins are modified by acetylation and the inhibition of HDAC activity can therefore affect various molecular processes. This broad effect on protein function may account for the pleiotropic anti-tumor responses elicited by HDACi that include induction of tumor cell apoptosis, cell cycle arrest, differentiation and senescence, modulation of immune responses and altered angiogenesis. The ability of HDACi to selectively induce tumor cells to undergo apoptosis is important for the therapeutic efficacy observed in pre-clinical models. Moreover, HDACi can augment the apoptotic effects of other anti-cancer agents that have diverse molecular targets. While HDACi are promising anti-cancer drugs, particularly given the scope to combine HDACi with other agents, identifying the key molecular events that determine the biological response of cells to HDACi treatment remains a challenge. Herein we focus on HDACi-induced apoptosis and discuss the various proteins and pathways that are affected by HDACi to mediate this programmed cell death response. In addition, we highlight the ability of HDACi to synergise with other anti-cancer agents to potently kill tumor cells and discuss the possible molecular processes that underpin the combination effect.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores de Histona Desacetilases , Animais , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Terapia Combinada , Sinergismo Farmacológico , Neoplasias Hematológicas/tratamento farmacológico , Histona Desacetilases/fisiologia , Humanos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Morte Celular/agonistas , Receptores de Morte Celular/fisiologia
4.
Nat Rev Cancer ; 8(10): 782-98, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18813321

RESUMO

Triggering of tumour cell apoptosis is the foundation of many cancer therapies. Death receptors of the tumour necrosis factor (TNF) superfamily have been largely characterized, as have the signals that are generated when these receptors are activated. TNF-related apoptosis-inducing ligand (TRAIL) receptors (TRAILR1 and TRAILR2) are promising targets for cancer therapy. Herein we review what is known about the molecular control of TRAIL-mediated apoptosis, the role of TRAIL in carcinogenesis and the potential therapeutic utility of recombinant TRAIL and agonistic antibodies against TRAILR1 and TRAILR2.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores de Morte Celular/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Camundongos , Neoplasias/genética , Receptores de Morte Celular/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/agonistas
5.
J Immunol ; 177(8): 5595-603, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17015747

RESUMO

Increased arginase I activity is associated with allergic disorders such as asthma. How arginase I contributes to and is regulated by allergic inflammatory processes remains unknown. CD4+ Th2 lymphocytes (Th2 cells) and IL-13 are two crucial immune regulators that use STAT6-dependent pathways to induce allergic airways inflammation and enhanced airways responsiveness to spasmogens (airways hyperresponsiveness (AHR)). This pathway is also used to activate arginase I in isolated cells and in hepatic infection with helminths. In the present study, we show that arginase I expression is also regulated in the lung in a STAT6-dependent manner by Th2-induced allergic inflammation or by IL-13 alone. IL-13-induced expression of arginase I correlated directly with increased synthesis of urea and with reduced synthesis of NO. Expression of arginase I, but not eosinophilia or mucus hypersecretion, temporally correlated with the development, persistence, and resolution of IL-13-induced AHR. Pharmacological supplementation with l-arginine or with NO donors amplified or attenuated IL-13-induced AHR, respectively. Moreover, inducing loss of function of arginase I specifically in the lung by using RNA interference abrogated the development of IL-13-induced AHR. These data suggest an important role for metabolism of l-arginine by arginase I in the modulation of IL-13-induced AHR and identify a potential pathway distal to cytokine receptor interactions for the control of IL-13-mediated bronchoconstriction in asthma.


Assuntos
Arginase/antagonistas & inibidores , Arginase/fisiologia , Hiper-Reatividade Brônquica/etiologia , Interleucina-13/fisiologia , Interferência de RNA , Animais , Arginase/genética , Arginase/metabolismo , Arginina/metabolismo , Asma/etiologia , Asma/metabolismo , Regulação Enzimológica da Expressão Gênica , Pulmão/enzimologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Doadores de Óxido Nítrico/metabolismo , Fator de Transcrição STAT6 , Células Th2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA