Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Curr Cancer Drug Targets ; 11(3): 254-84, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21247382

RESUMO

The proteasome has emerged as an important clinically relevant target for the treatment of hematologic malignancies. Since the Food and Drug Administration approved the first-in-class proteasome inhibitor bortezomib (Velcade) for the treatment of relapsed/refractory multiple myeloma (MM) and mantle cell lymphoma, it has become clear that new inhibitors are needed that have a better therapeutic ratio, can overcome inherent and acquired bortezomib resistance and exhibit broader anti-cancer activities. Marizomib (NPI-0052; salinosporamide A) is a structurally and pharmacologically unique ß-lactone-γ-lactam proteasome inhibitor that may fulfill these unmet needs. The potent and sustained inhibition of all three proteolytic activities of the proteasome by marizomib has inspired extensive preclinical evaluation in a variety of hematologic and solid tumor models, where it is efficacious as a single agent and in combination with biologics, chemotherapeutics and targeted therapeutic agents. Specifically, marizomib has been evaluated in models for multiple myeloma, mantle cell lymphoma, Waldenstrom's macroglobulinemia, chronic and acute lymphocytic leukemia, as well as glioma, colorectal and pancreatic cancer models, and has exhibited synergistic activities in tumor models in combination with bortezomib, the immunomodulatory agent lenalidomide (Revlimid), and various histone deacetylase inhibitors. These and other studies provided the framework for ongoing clinical trials in patients with MM, lymphomas, leukemias and solid tumors, including those who have failed bortezomib treatment, as well as in patients with diagnoses where other proteasome inhibitors have not demonstrated significant efficacy. This review captures the remarkable translational studies and contributions from many collaborators that have advanced marizomib from seabed to bench to bedside.


Assuntos
Antineoplásicos/uso terapêutico , Lactonas/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Inibidores de Proteassoma , Pirróis/uso terapêutico , Animais , Avaliação Pré-Clínica de Medicamentos , Humanos , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo
2.
Crit Rev Oncog ; 16(3-4): 261-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22248059

RESUMO

The transcription factor Yin Yang (YY) 1 has been reported to be overexpressed in several tumor types and plays a role in both the progression of the disease as well as the maintenance of tumor cell resistance to cell death by cytotoxic drugs. YY1 also has been reported to be a prognostic factor for several cancers and was proposed to be a therapeutic target. The expression, function, and role of YY1 in the pathogenesis of hematologic malignancies are summarized briefly herein. Data are represented for B non-Hodgkin lymphoma, AIDS-related lymphoma, multiple myeloma, and children's acute lymphocytic leukemia.


Assuntos
Neoplasias Hematológicas/patologia , Fator de Transcrição YY1/fisiologia , Neoplasias Hematológicas/genética , Humanos , Fator de Transcrição YY1/genética
3.
Leukemia ; 23(1): 59-70, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18946492

RESUMO

The human transferrin receptor (hTfR) is a target for cancer immunotherapy due to its overexpression on the surface of cancer cells. We previously developed an antibody-avidin fusion protein that targets hTfR (anti-hTfR IgG3-Av) and exhibits intrinsic cytotoxicity against certain malignant cells. Gambogic acid (GA), a drug that also binds hTfR, induces cytotoxicity in several malignant cell lines. We now report that anti-hTfR IgG3-Av and GA induce cytotoxicity in a new broader panel of hematopoietic malignant cell lines. Our results show that the effect of anti-hTfR IgG3-Av is iron-dependent whereas that of GA is iron-independent in all cells tested. In addition, we observed that GA exerts a TfR-independent cytotoxicity. We also found that GA increases the generation of reactive oxygen species that may play a role in the cytotoxicity induced by this drug. Additive cytotoxicity was observed by simultaneous combination treatment with these drugs and synergy by using anti-hTfR IgG3-Av as a chemosensitizing agent. In addition, we found a concentration of GA that is toxic to malignant hematopoietic cells but not to human hematopoietic progenitor cells. Our results suggest that these two compounds may be effective, alone or in combination, for the treatment of human hematopoietic malignancies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Hematológicas/tratamento farmacológico , Imunoglobulina G/farmacologia , Receptores da Transferrina/imunologia , Xantonas/farmacologia , Avidina , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Humanos , Imunoglobulina G/uso terapêutico , Ferro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Xantonas/uso terapêutico
4.
Oncogene ; 26(25): 3629-36, 2007 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-17530016

RESUMO

Rituximab (chimeric anti-CD20 monoclonal antibody) is the first Food and Drug Administration approved antitumor antibody and is used in the treatment of B-non-Hodgkin's lymphoma (B-NHL). It is used as single monotherapy or in combination with chemotherapy and has improved the treatment outcome of patients with B-NHL. The in vivo mechanisms of rituximab-mediated antitumor effects include antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cell cytotoxicity (CDC), growth-inhibition and apoptosis. A subset of patients does not initially respond to rituximab and several responsive patients develop resistance to further rituximab treatment. The mechanism of rituximab unresponsiveness is not known. Besides the above-postulated mechanisms, rituximab has been shown to trigger the cells via CD-20. Studies performed with B-NHL cell lines as model systems revealed several novel mechanisms of rituximab-mediated effects that are involved in chemo/immunosensitization and the development of resistance to rituximab. Rituximab has been shown to inhibit the p38 mitogen-activated protein kinase, nuclear factor-kappaB (NF-kappaB), extracellular signal-regulated kinase 1/2 (ERK 1/2) and AKT antiapoptotic survival pathways, all of which result in upregulation of phosphatase and tensin homolog deleted on chromosome ten and Raf kinase inhibitor protein and in the downregulation of antiapoptotic gene products (particularly Bcl-2, Bcl-(xL) and Mcl-1), and resulting in chemo/immunosensitization. Further, rituximab treatment inhibits the overexpressed transcription repressor Yin Yang 1 (YY1), which negatively regulates Fas and DR5 expression and its inhibition leads to sensitization to Fas ligand and tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Rituximab-resistant clones were generated as model to examine the mechanism of in vivo rituximab unresponsiveness. These clones showed reduced expression of CD20 and hyperactivation of the above antiapoptotic signaling pathways and failure of rituximab to trigger the cells leading to inhibition of ADCC, CDC and chemo/immunosensitization. Interference with the hyperactivated pathways with various pharmacological and proteasome inhibitors reversed resistance. Furthermore, the above findings have identified several gene products that can serve as new prognostic/diagnostic biomarkers as well as targets for therapeutic intervention in B-NHL.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Imunoterapia , Linfoma de Células B/patologia , Linfoma de Células B/terapia , Anticorpos Monoclonais Murinos , Humanos , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo , Prognóstico , Rituximab
5.
Oncogene ; 25(8): 1125-42, 2006 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-16314846

RESUMO

The ubiquitous transcription factor Yin Yang 1 (YY1) is known to have a fundamental role in normal biologic processes such as embryogenesis, differentiation, replication, and cellular proliferation. YY1 exerts its effects on genes involved in these processes via its ability to initiate, activate, or repress transcription depending upon the context in which it binds. Mechanisms of action include direct activation or repression, indirect activation or repression via cofactor recruitment, or activation or repression by disruption of binding sites or conformational DNA changes. YY1 activity is regulated by transcription factors and cytoplasmic proteins that have been shown to abrogate or completely inhibit YY1-mediated activation or repression; however, these mechanisms have not yet been fully elucidated. Since expression and function of YY1 are known to be intimately associated with progression through phases of the cell cycle, the physiologic significance of YY1 activity has recently been applied to models of tumor biology. The majority of the data are consistent with the hypothesis that YY1 overexpression and/or activation is associated with unchecked cellular proliferation, resistance to apoptotic stimuli, tumorigenesis and metastatic potential. Studies involving hematopoetic tumors, epithelial-based tumors, endocrine organ malignancies, hepatocellular carcinoma, and retinoblastoma support this hypothesis. Molecular mechanisms that have been investigated include YY1-mediated downregulation of p53 activity, interference with poly-ADP-ribose polymerase, alteration in c-myc and nuclear factor-kappa B (NF-kappaB) expression, regulation of death genes and gene products, and differential YY1 binding in the presence of inflammatory mediators. Further, recent findings implicate YY1 in the regulation of tumor cell resistance to chemotherapeutics and immune-mediated apoptotic stimuli. Taken together, these findings provide strong support of the hypothesis that YY1, in addition to its regulatory roles in normal biologic processes, may possess the potential to act as an initiator of tumorigenesis and may thus serve as both a diagnostic and prognostic tumor marker; furthermore, it may provide an effective target for antitumor chemotherapy and/or immunotherapy.


Assuntos
Neoplasias , Fator de Transcrição YY1 , Animais , Sequência de Bases , Humanos , Oncologia , Dados de Sequência Molecular , Neoplasias/diagnóstico , Neoplasias/metabolismo , Neoplasias/terapia , Fator de Transcrição YY1/química , Fator de Transcrição YY1/metabolismo , Fator de Transcrição YY1/uso terapêutico
6.
Biomarkers ; 10 Suppl 1: S69-76, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16298914

RESUMO

PRIMARY OBJECTIVE: The primary objective is to delineate the potential utility of cancer biomarkers that correlate and predict response to immunotherapy in cancer patients who are refractory to conventional therapeutics. Unlike significant development of biomarkers that predict response to chemotherapy, very few biomarkers have been developed to predict the response to immunotherapy. MAIN OUTCOMES AND RESULTS: This article describes briefly the importance of characterizing and validating biomarkers for immunotherapy. A few examples have been provided, such as the transcription factor NF-kappaB, the transcription repressor Yin-Yang 1 (YY1), the pro-apoptotic gene product (Smac/DIABLO) and the circulating Fas and Fas ligand. These biomarkers have been determined to be of prognostic significance in different cancers. CONCLUSIONS: Immunotherapy is considered as an alternative therapy in the treatment of cancer patients who are refractory to chemotherapy/radiation/hormonal therapies. Cross-resistance to apoptosis develops between cancer cells that are resistant to conventional therapeutics and immunotherapy. Therefore, it is important to develop biomarkers that will determine patient response to immunotherapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Biomarcadores/metabolismo , Imunoterapia/métodos , Neoplasias/terapia , Animais , Proteínas Reguladoras de Apoptose , Proteína Ligante Fas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Neoplasias/diagnóstico , Resultado do Tratamento , Fatores de Necrose Tumoral/metabolismo , Fator de Transcrição YY1/metabolismo , Receptor fas/metabolismo
7.
J Immunol ; 167(1): 75-81, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11418634

RESUMO

NO has been increasingly implicated in control of the transcriptional machinery and serves as an intracellular second messenger to modify gene expression. We have demonstrated that NO up-regulated Fas receptor expression in ovarian carcinoma cell lines, albeit the mechanism involved is not known. Thus, we hypothesized that NO, directly or indirectly, may modify the transcriptional machinery that is responsible for the increased expression of the Fas gene. We examined the effect of NO on Fas gene expression using a Fas promoter-driven luciferase reporter system. Transient transfection of AD10 cells with pGL-3-FasP demonstrated that the IFN-gamma-dependent NO generation increases the trans-activation of the Fas promoter, and this increase was blocked by the NOS inhibitor (N(G)-monomethyl-L-arginine), but could be restored by the addition of the NO donor S-nitroso-N-acetylpenicillamine. Systematic deletion of the Fas promoter revealed that the functional region responsible for the NO-mediated effect was located at the silencer region, suggesting that NO may be responsible for the disruption of a repressor mechanism. We demonstrate that NO up-regulates the expression of the Fas receptor on AD10 cells via the specific inactivation of the transcription repressor yin-yang 1 DNA binding activity to the silencer region of the Fas promoter. These findings reveal a new mechanism of NO-mediated gene regulation by interfering with a repressor transcription factor at the silencer region of the Fas promoter.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/imunologia , Inativação Gênica/imunologia , Óxido Nítrico/fisiologia , Regiões Promotoras Genéticas/imunologia , Proteínas Repressoras/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Regulação para Cima/imunologia , Receptor fas/genética , Sequência de Bases , DNA/antagonistas & inibidores , DNA/metabolismo , Proteínas de Ligação a DNA/biossíntese , Fatores de Ligação de DNA Eritroide Específicos , Humanos , Interferon gama/fisiologia , Dados de Sequência Molecular , Ligação Proteica/genética , Ligação Proteica/imunologia , RNA Mensageiro/biossíntese , Proteínas Repressoras/biossíntese , Proteínas Repressoras/metabolismo , Elementos de Resposta/imunologia , Fatores de Transcrição/biossíntese , Células Tumorais Cultivadas , Regulação para Cima/genética , Fator de Transcrição YY1 , Receptor fas/biossíntese
8.
J Immunol ; 117(6): 2204-8, 1976 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-792342

RESUMO

Mice of 1.5, 9, 22, and 31 to 32 months of age were injected with the thymus-dependent antigen, TNP-SRC, or the thymus-independent antigen, TNP-SRC, TNP-MRC. The anti-SRC and TNP immune responses to TNP-SRC were markedly reduced in older mice, whereas the anti-TNP response to the TNP-MRC showed no substantial decline. Young mice produced higher anti-TNP plaque-forming cell responses after injection of TNP-SRC than after TNP-MRC, whereas in older mice the reverse obtained. Old mice but not young mice displayed a high anti-SRC cross-reactive response after injection of TNP-MRC. The avidity of anti-TNP antibody of young mice immunized with TNP-SRC was higher than that following immunization with TNP-MRC, whereas the avidities of anti-TNP antibodies from old mice injected with these two reagents were the same. Those individual mice which showed a poorly regulated immune response also displayed an autologous anti-MRC plaque-forming cell response after injection of either TNP-SRC or TNP-MRC. It is suggested that mechanisms mediated by suppressor T cells may be responsible for regulating the autoimmune response to modified self antigens, and that these are severely impaired in age individuals.


Assuntos
Envelhecimento , Autoanticorpos , Eritrócitos/imunologia , Nitrobenzenos/imunologia , Trinitrobenzenos/imunologia , Animais , Formação de Anticorpos , Bromelaínas/farmacologia , Relação Dose-Resposta Imunológica , Técnica de Placa Hemolítica , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Perissodáctilos , Ovinos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA