Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 139(18): 2770-2781, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35226739

RESUMO

Small ubiquitin-like modifier (SUMO) is a member of a ubiquitin-like protein superfamily. SUMOylation is a reversible posttranslational modification that has been implicated in the regulation of various cellular processes including inflammatory responses and expression of type 1 interferons (IFN1). In this report, we have explored the activity of the selective small molecule SUMOylation inhibitor subasumstat (TAK-981) in promoting antitumor innate immune responses. We demonstrate that treatment with TAK-981 results in IFN1-dependent macrophage and natural killer (NK) cell activation, promoting macrophage phagocytosis and NK cell cytotoxicity in ex vivo assays. Furthermore, pretreatment with TAK-981 enhanced macrophage phagocytosis or NK cell cytotoxicity against CD20+ target cells in combination with the anti-CD20 antibody rituximab. In vivo studies demonstrated enhanced antitumor activity of TAK-981 and rituximab in CD20+ lymphoma xenograft models. Combination of TAK-981 with anti-CD38 antibody daratumumab also resulted in enhanced antitumor activity. TAK-981 is currently being studied in phase 1 clinical trials (#NCT03648372, #NCT04074330, #NCT04776018, and #NCT04381650; www.clinicaltrials.gov) for the treatment of patients with lymphomas and solid tumors.


Assuntos
Linfoma , Sumoilação , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD20 , Linhagem Celular Tumoral , Humanos , Células Matadoras Naturais , Linfoma/tratamento farmacológico , Linfoma/metabolismo , Macrófagos/metabolismo , Rituximab/metabolismo , Rituximab/farmacologia , Rituximab/uso terapêutico
2.
Blood ; 117(17): 4658-66, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21378271

RESUMO

CD36 plays a critical role in the inhibition of angiogenesis through binding to the type 1 repeats of thrombospondin-1 (TSP-1) and activating Fyn tyrosine kinase and MAPK pathways. Here, we reveal a novel association of CD36 with VEGFR-2 and spleen tyrosine kinase (Syk). We also address the correlation between the expression of CD36 and Syk by demonstrating that overexpression of CD36 in HUVECs up-regulates endogenous Syk expression. We also define a new role for TSP-1 and CD36 in the activation of the VEGFR-2 signaling pathway that requires Syk. Our findings also identify a role for Syk as a stimulator of VEGF-A-induced angiogenesis by increasing phosphorylation of Y1175 in VEGFR-2, which is a major tyrosine for promoting VEGF-A-induced endothelial cell migration. Together, these studies introduce a new signaling pathway for TSP-1, CD36, and Syk, and address the role of these proteins in regulating the angiogenic switch.


Assuntos
Antígenos CD36/metabolismo , Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Trombospondina 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Movimento Celular/fisiologia , Células Cultivadas , Células Endoteliais/citologia , Humanos , Neovascularização Fisiológica/fisiologia , Fosforilação/fisiologia , Quinase Syk , Veias Umbilicais/citologia , Regulação para Cima/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Sci Transl Med ; 13(611): eaba7791, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34524860

RESUMO

SUMOylation, the covalent conjugation of small ubiquitin-like modifier (SUMO) proteins to protein substrates, has been reported to suppress type I interferon (IFN1) responses. TAK-981, a selective small-molecule inhibitor of SUMOylation, pharmacologically reactivates IFN1 signaling and immune responses against cancers. In vivo treatment of wild-type mice with TAK-981 up-regulated IFN1 gene expression in blood cells and splenocytes. Ex vivo treatment of mouse and human dendritic cells promoted their IFN1-dependent activation, and vaccination studies in mice demonstrated stimulation of antigen cross-presentation and T cell priming in vivo. TAK-981 also directly stimulated T cell activation, driving enhanced T cell sensitivity and response to antigen ex vivo. Consistent with these observations, TAK-981 inhibited growth of syngeneic A20 and MC38 tumors in mice, dependent upon IFN1 signaling and CD8+ T cells, and associated with increased intratumoral T and natural killer cell number and activation. Combination of TAK-981 with anti-PD1 or anti-CTLA4 antibodies improved the survival of mice bearing syngeneic CT26 and MC38 tumors. In conclusion, TAK-981 is a first-in-class SUMOylation inhibitor that promotes antitumor immune responses through activation of IFN1 signaling. TAK-981 is currently being studied in phase 1 clinical trials (NCT03648372, NCT04074330, NCT04776018, and NCT04381650) for the treatment of patients with solid tumors and lymphomas.


Assuntos
Imunidade , Sumoilação
4.
J Med Chem ; 64(5): 2501-2520, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33631934

RESUMO

SUMOylation is a reversible post-translational modification that regulates protein function through covalent attachment of small ubiquitin-like modifier (SUMO) proteins. The process of SUMOylating proteins involves an enzymatic cascade, the first step of which entails the activation of a SUMO protein through an ATP-dependent process catalyzed by SUMO-activating enzyme (SAE). Here, we describe the identification of TAK-981, a mechanism-based inhibitor of SAE which forms a SUMO-TAK-981 adduct as the inhibitory species within the enzyme catalytic site. Optimization of selectivity against related enzymes as well as enhancement of mean residence time of the adduct were critical to the identification of compounds with potent cellular pathway inhibition and ultimately a prolonged pharmacodynamic effect and efficacy in preclinical tumor models, culminating in the identification of the clinical molecule TAK-981.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Ácidos Sulfônicos/uso terapêutico , Sumoilação/efeitos dos fármacos , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Humanos , Camundongos , Estrutura Molecular , Ligação Proteica , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Relação Estrutura-Atividade , Ácidos Sulfônicos/síntese química , Ácidos Sulfônicos/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Methods Enzymol ; 446: 315-31, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18603131

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family of cytokines. TRAIL has gained much attention because of its ability to preferentially kill tumor cells with no apparent toxic side effects. Recently, different TRAIL receptor agonists, including TRAIL itself and various agonistic monoclonal antibodies against the two apoptosis-inducing human TRAIL receptors, have been developed as novel cancer therapeutics and are currently under investigation in clinical trials. However, the mechanisms by which TRAIL mediates its selective antineoplastic activity are still not well understood. In addition to playing a role in cancer immune surveillance and tumor suppression, TRAIL has been associated with immune homeostasis, inflammatory diseases, and autoimmunity. In light of the multifunctional role of TRAIL in mediating various pathologic conditions and the potential benefits of TRAIL-based therapies, the study of the physiologic significance of TRAIL is of great importance. Here, we describe a syngeneic system for the characterization of the in vivo function of TRAIL. By use of this model, in which the full-length murine TRAIL protein is overexpressed in the hematopoietic cells of wild-type mice, the in vivo tumoricidal activity of TRAIL overexpression can be studied on syngeneic murine tumor cell challenge, and the potential toxicity of TRAIL protein to normal tissues can also be analyzed.


Assuntos
Transplante de Medula Óssea/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Transplante de Medula Óssea/métodos , Imuno-Histoquímica/métodos , Camundongos , Retroviridae/genética , Baço/patologia , Transdução Genética/métodos
6.
Adv Exp Med Biol ; 615: 47-79, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18437891

RESUMO

Apoptosis is a cell suicide program that plays a critical role in development and tissue homeostasis. The ability of cancer cells to evade this programmed cell death (PCD) is a major characteristic that enables their uncontrolled growth. The efficiency of chemotherapy in killing such cells depends on the successful induction of apoptosis, since defects in apoptosis signaling are a major cause of drug resistance. Over the past decades, much progress has been made in our understanding of apoptotic signaling pathways and their dysregulation in cancer progression and therapy. These advances have provided new molecular targets for proapoptotic cancer therapies that have recently been used in drug development. While most of those therapies are still at the preclinical stage, some of them have shown much promise in the clinic. Here, we review our current knowledge of apoptosis regulation in cancer progression and therapy, as well as the new molecular targeted molecules that are being developed to reinstate cancer cell death.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos , Animais , Progressão da Doença , Humanos , Neoplasias/metabolismo
7.
Cancer Res ; 66(12): 6304-11, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778207

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family of cytokines and has been shown to induce cell death in many types of tumor and transformed cells but not in normal cells. This tumor-selective property has made TRAIL a promising candidate for the development of cancer therapy. However, safety issues are a concern because certain preparations of recombinant TRAIL protein were reported to induce toxicity in normal human hepatocytes in culture. In addition, previous studies on tumor selectivity of exogenous TRAIL protein were carried out in xenograft models, which do not directly address the tumor selectivity issue. It was not known whether exogenous or overexpression of TRAIL in a syngeneic system could induce tumor cell death while leaving normal tissue cells unharmed. Thus, the tumor selectivity of TRAIL-induced apoptosis remains to be further characterized. In our study, we established mice that overexpress TRAIL by retroviral-mediated gene transfer in bone marrow cells followed by bone marrow transplantation. Our results show that TRAIL overexpression is not toxic to normal tissues, as analyzed by hematologic and histologic analyses of tissue samples from TRAIL-transduced mice. We show for the first time that TRAIL overexpression in hematopoietic cells leads to significant inhibition of syngeneic tumor growth in certain tumor lines. This approach may be used further to identify important molecules that regulate the sensitivity of tumor cells to TRAIL-induced cell death in vivo.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea/métodos , Terapia Genética/métodos , Células-Tronco Hematopoéticas/fisiologia , Glicoproteínas de Membrana/genética , Neoplasias Experimentais/terapia , Fator de Necrose Tumoral alfa/genética , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/biossíntese , Células da Medula Óssea/metabolismo , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Células-Tronco Hematopoéticas/metabolismo , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Retroviridae/genética , Ligante Indutor de Apoptose Relacionado a TNF , Transdução Genética , Transfecção , Fator de Necrose Tumoral alfa/biossíntese
9.
Cancer Res ; 69(9): 3856-65, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19366809

RESUMO

As tumor development relies on a coordination of angiogenesis and tumor growth, an efficient antitumor strategy should target both the tumor and its associated vessels. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in a tumor-selective manner. Additionally, thrombospondin-1, a naturally occurring inhibitor of angiogenesis, and a recombinant protein containing functional domains of thrombospondin-1, 3TSR, have been shown to be necessary and sufficient to inhibit tumor angiogenesis. Here, we show that a combination of a TRAIL receptor 2 agonist antibody, Lexatumumab, and 3TSR results in a significantly enhanced and durable tumor inhibition. We further observed that 3TSR induces apoptosis in primary endothelial cells by up-regulating the expression of TRAIL receptors 1 and 2 in a CD36 and Jun NH(2)-terminal kinase-dependent manner leading to the activation of both intrinsic and extrinsic apoptotic machineries. The modulation of these pathways is critical for 3TSR-induced apoptosis as disrupting either via specific inhibitors reduced apoptosis. Moreover, 3TSR attenuates the Akt survival pathway. These studies indicate that 3TSR plays a critical role in regulating the proapoptotic signaling pathways that control growth and death in endothelial cells and that a combination of TRAIL and 3TSR acts as a double hit against tumor and tumor-associated vessels.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 8/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Ativação Enzimática , Feminino , Células HCT116 , Humanos , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Fragmentos de Peptídeos/administração & dosagem , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Proteínas Recombinantes/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Trombospondinas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 69(16): 6546-55, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19654305

RESUMO

BCR-ABL plays an essential role in the pathogenesis of chronic myeloid leukemia (CML) and some cases of acute lymphocytic leukemia (ALL). Although ABL kinase inhibitors have shown great promise in the treatment of CML, the persistence of residual disease and the occurrence of resistance have prompted investigations into the molecular effectors of BCR-ABL. Here, we show that BCR-ABL stimulates the proteasome-dependent degradation of members of the forkhead family of tumor suppressors in vitro, in an in vivo animal model, and in samples from patients with BCR-ABL-positive CML or ALL. As several downstream mediators of BCR-ABL are regulated by the proteasome degradation pathway, we also show that inhibition of this pathway, using bortezomib, causes regression of CML-like disease. Bortezomib treatment led to inhibition of BCR-ABL-induced suppression of FoxO proteins and their proapoptotic targets, tumor necrosis factor-related apoptosis-inducing ligand and BIM, thereby providing novel insights into the molecular effects of proteasome inhibitor therapy. We additionally show sensitivity of imatinib-resistant BCR-ABL T315I cells to bortezomib. Our data delineate the involvement of FoxO proteins in BCR-ABL-induced evasion of apoptosis and provide evidence that bortezomib is a candidate therapeutic in the treatment of BCR-ABL-induced leukemia.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Cisteína Proteinase/uso terapêutico , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Fusão bcr-abl/fisiologia , Leucemia/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/genética , Benzamidas , Ácidos Borônicos/farmacologia , Ácidos Borônicos/uso terapêutico , Bortezomib , Inibidores de Cisteína Proteinase/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Fatores de Transcrição Forkhead/fisiologia , Proteínas de Fusão bcr-abl/genética , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Leucemia/genética , Leucemia/patologia , Camundongos , Camundongos Nus , Piperazinas/farmacologia , Inibidores de Proteassoma , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Pirimidinas/farmacologia , Indução de Remissão , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Vaccine ; 22(9-10): 1199-205, 2004 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-15003648

RESUMO

Vaccination with plasmids encoding an antigen of interest (DNA vaccination) is a new strategy to achieve effective immunization against many agents. DNA vaccination can be ameliorated by co-administration of plasmids encoding a cytokine. Thus far, only plasmids encoding soluble cytokines have been used for this purpose. However, these plasmids can induce release of cytokines into the circulation and could potentially cause many undesirable effects. We undertook this study to determine whether membrane-bound cytokines, which would restrict their localization at the site of administration, can act as immunoadjuvants. We and others have previously shown that plasmids encoding soluble IL-4 and IL-12 are effective adjuvants for DNA vaccination. In this study, we demonstrate that DNA co-vaccination with membrane-bound IL-4 (mbIL-4) or membrane-bound IL-12 (mbIL-12) both enhance anti-CEA immunity, as detected by in vitro and in vivo assays. Mice co-injected with plasmids encoding CEA and either type of membrane-bound cytokine rejected transplanted CEA-positive tumor cells strongly. Notably, unlike secreted IL-4, mbIL-4 was the most effective adjuvant for anti-tumor immunity. This study demonstrates that membrane-bound cytokines are suitable adjuvants for DNA vaccination.


Assuntos
Antígeno Carcinoembrionário/genética , Interleucina-12/genética , Interleucina-4/genética , Plasmídeos/genética , Vacinas de DNA/genética , Adjuvantes Imunológicos , Animais , Células COS , Carcinoma Pulmonar de Lewis/metabolismo , Chlorocebus aethiops , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Baço/citologia
12.
J Biol Chem ; 279(53): 55594-601, 2004 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-15485835

RESUMO

Here we show a novel mechanism by which FLICE-like inhibitory protein (c-FLIP) regulates apoptosis induced by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and one of its receptors, DR5. c-FLIP is a critical regulator of the TNF family of cytokine receptor signaling. c-FLIP has been postulated to prevent formation of the competent death-inducing signaling complex (DISC) in a ligand-dependent manner, through its interaction with FADD and/or caspase-8. In order to identify regulators of TRAIL function, we used the intracellular death domain (DD) of DR5 as a target to screen a phage-displayed combinatorial peptide library. The DD of DR5 selected from the library a peptide that showed sequence similarity to a stretch of amino acids in the C terminus of c-FLIP(L). The phage-displayed peptide selectively interacted with the DD of DR5 in in vitro binding assays. Similarly, full-length c-FLIP (c-FLIP(L)) and the C-terminal p12 domain of c-FLIP interacted with DR5 both in in vitro pull-down assays and in mammalian cells. This interaction was independent of TRAIL. To the contrary, TRAIL treatment released c-FLIP(L) from DR5, permitting the recruitment of FADD to the active DR5 signaling complex. By employing FADD-deficient Jurkat cells, we demonstrate that DR5 and c-FLIP(L) interact in a FADD-independent manner. Moreover, we show that a cellular membrane permeable version of the peptide corresponding to the DR5 binding domain of c-FLIP induces apoptosis in mammalian cells. Taken together, these findings indicate that c-FLIP interacts with the DD of DR5, thus preventing death (L)signaling by DR5 prior to the formation of an active DISC. Because TRAIL and DR5 are ubiquitously expressed, the interaction of c-FLIP(L) and DR5 indicates a mechanism by which tumor selective apoptosis can be achieved through protecting normal cells from undergoing death receptor-induced apoptosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores do Fator de Necrose Tumoral/química , Proteínas Adaptadoras de Transdução de Sinal/química , Alanina/química , Apoptose , Proteínas Reguladoras de Apoptose , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Caspase 8 , Caspases/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Proteína de Domínio de Morte Associada a Fas , Glutationa Transferase/metabolismo , Humanos , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/química , Células Jurkat , Ligantes , Glicoproteínas de Membrana/metabolismo , Microscopia de Fluorescência , Biblioteca de Peptídeos , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Ligante Indutor de Apoptose Relacionado a TNF , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA