Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 28(8): 1540-1548, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35140121

RESUMEN

PURPOSE: Dual inhibition of glucose and glutamine metabolism results in synergistic anticancer effects in solid tumor models. Telaglenastat, an investigational, small-molecule, glutaminase inhibitor, exhibits modest single-agent activity in renal cell carcinoma (RCC) patients. This phase Ib trial evaluated telaglenastat plus cabozantinib or everolimus, agents known to impair glucose metabolism in patients with metastatic RCC (mRCC). PATIENTS AND METHODS: mRCC patients received escalating doses of telaglenastat [400-800 mg per os (p.o.) twice daily] in a 3 + 3 design, plus either everolimus (10 mg daily p.o.; TelaE) or cabozantinib (60 mg daily p.o.; TelaC). Tumor response (RECISTv1.1) was assessed every 8 weeks. Endpoints included safety (primary) and antitumor activity. RESULTS: Twenty-seven patients received TelaE, 13 received TelaC, with median 2 and 3 prior therapies, respectively. Treatment-related adverse events were mostly grades 1 to 2, most common including decreased appetite, anemia, elevated transaminases, and diarrhea with TelaE, and diarrhea, decreased appetite, elevated transaminases, and fatigue with TelaC. One dose-limiting toxicity occurred per cohort: grade 3 pruritic rash with TelaE and thrombocytopenia with TelaC. No maximum tolerated dose (MTD) was reached for either combination, leading to a recommended phase II dose of 800-mg telaglenastat twice daily with standard doses of E or C. TelaE disease control rate (DCR; response rate + stable disease) was 95.2% [20/21, including 1 partial response (PR)] among 21 patients with clear cell histology and 66.7% (2/3) for papillary. TelaC DCR was 100% (12/12) for both histologies [5/10 PRs as best response (3 confirmed) in clear cell]. CONCLUSIONS: TelaE and TelaC showed encouraging clinical activity and tolerability in heavily pretreated mRCC patients.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Anilidas , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Diarrea/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Everolimus , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Piridinas , Transaminasas
2.
Clin Cancer Res ; 27(18): 4994-5003, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34285061

RESUMEN

PURPOSE: Glutamine is a critical fuel for solid tumors. Interference with glutamine metabolism is deleterious to neoplasia in preclinical models. A phase I study of the oral, first-in-class, glutaminase (GLS) inhibitor telaglenastat was conducted in treatment-refractory solid tumor patients to define recommended phase II dose (RP2D) and evaluate safety, pharmacokinetics (PK), pharmacodynamics (PD), and antitumor activity. PATIENTS AND METHODS: Dose escalation by 3 + 3 design was followed by exploratory tumor-/biomarker-specific cohorts. RESULTS: Among 120 patients, fatigue (23%) and nausea (19%) were the most common toxicity. Maximum tolerated dose was not reached. Correlative analysis indicated >90% GLS inhibition in platelets at plasma exposures >300 nmol/L, >75% tumoral GLS inhibition, and significant increase in circulating glutamine. RP2D was defined at 800 mg twice-daily. Disease control rate (DCR) was 43% across expansion cohorts (overall response rate 5%, DCR 50% in renal cell carcinoma). CONCLUSIONS: Telaglenastat is safe, with a favorable PK/PD profile and signal of antitumor activity, supporting further clinical development.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Neoplasias , Inhibidores Enzimáticos , Humanos , Dosis Máxima Tolerada , Náusea , Neoplasias/tratamiento farmacológico
3.
J Immunother Cancer ; 5(1): 101, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29254508

RESUMEN

BACKGROUND: Myeloid cells are an abundant leukocyte in many types of tumors and contribute to immune evasion. Expression of the enzyme arginase 1 (Arg1) is a defining feature of immunosuppressive myeloid cells and leads to depletion of L-arginine, a nutrient required for T cell and natural killer (NK) cell proliferation. Here we use CB-1158, a potent and orally-bioavailable small-molecule inhibitor of arginase, to investigate the role of Arg1 in regulating anti-tumor immunity. METHODS: CB-1158 was tested for the ability to block myeloid cell-mediated inhibition of T cell proliferation in vitro, and for tumor growth inhibition in syngeneic mouse models of cancer as a single agent and in combination with other therapies. Tumors from animals treated with CB-1158 were profiled for changes in immune cell subsets, expression of immune-related genes, and cytokines. Human tumor tissue microarrays were probed for Arg1 expression by immunohistochemistry and immunofluorescence. Cancer patient plasma samples were assessed for Arg1 protein and L-arginine by ELISA and mass spectrometry, respectively. RESULTS: CB-1158 blocked myeloid cell-mediated suppression of T cell proliferation in vitro and reduced tumor growth in multiple mouse models of cancer, as a single agent and in combination with checkpoint blockade, adoptive T cell therapy, adoptive NK cell therapy, and the chemotherapy agent gemcitabine. Profiling of the tumor microenvironment revealed that CB-1158 increased tumor-infiltrating CD8+ T cells and NK cells, inflammatory cytokines, and expression of interferon-inducible genes. Patient tumor samples from multiple histologies expressed an abundance of tumor-infiltrating Arg1+ myeloid cells. Plasma samples from cancer patients exhibited elevated Arg1 and reduced L-arginine compared to healthy volunteers. CONCLUSIONS: These results demonstrate that Arg1 is a key mediator of immune suppression and that inhibiting Arg1 with CB-1158 shifts the immune landscape toward a pro-inflammatory environment, blunting myeloid cell-mediated immune evasion and reducing tumor growth. Furthermore, our results suggest that arginase blockade by CB-1158 may be an effective therapy in multiple types of cancer and combining CB-1158 with standard-of-care chemotherapy or other immunotherapies may yield improved clinical responses.


Asunto(s)
Arginasa/metabolismo , Células Mieloides/citología , Neoplasias/tratamiento farmacológico , Pirrolidinas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Animales , Arginasa/antagonistas & inhibidores , Arginina/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Células Hep G2 , Humanos , Células K562 , Masculino , Ratones , Células Mieloides/efectos de los fármacos , Células Mieloides/enzimología , Neoplasias/inmunología , Neoplasias/metabolismo , Pirrolidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Br J Haematol ; 173(6): 884-95, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27071340

RESUMEN

While proteasome inhibition is a validated therapeutic approach for multiple myeloma (MM), inhibition of individual constitutive proteasome (c20S) and immunoproteasome (i20S) subunits has not been fully explored owing to a lack of effective tools. We utilized the novel proteasome constitutive/immunoproteasome subunit enzyme-linked immunosorbent (ProCISE) assay to quantify proteasome subunit occupancy in samples from five phase I/II and II trials before and after treatment with the proteasome inhibitor carfilzomib. Following the first carfilzomib dose (15-56 mg/m(2) ), dose-dependent inhibition of c20S and i20S chymotrypsin-like active sites was observed [whole blood: ≥67%; peripheral blood mononuclear cells (PBMCs): ≥75%]. A similar inhibition profile was observed in bone marrow-derived CD138(+) tumour cells. Carfilzomib-induced proteasome inhibition was durable, with minimal recovery in PBMCs after 24 h but near-complete recovery between cycles. Importantly, the ProCISE assay can be used to quantify occupancy of individual c20S and i20S subunits. We observed a relationship between MM patient response (n = 29), carfilzomib dose and occupancy of multiple i20S subunits, where greater occupancy was associated with an increased likelihood of achieving a clinical response at higher doses. ProCISE represents a new tool for measuring proteasome inhibitor activity in clinical trials and relating drug action to patient outcomes.


Asunto(s)
Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Antineoplásicos/uso terapéutico , Médula Ósea/patología , Relación Dosis-Respuesta a Droga , Humanos , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Inducción de Remisión , Células Tumorales Cultivadas
5.
Mol Cancer Ther ; 13(4): 890-901, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24523301

RESUMEN

Glutamine serves as an important source of energy and building blocks for many tumor cells. The first step in glutamine utilization is its conversion to glutamate by the mitochondrial enzyme glutaminase. CB-839 is a potent, selective, and orally bioavailable inhibitor of both splice variants of glutaminase (KGA and GAC). CB-839 had antiproliferative activity in a triple-negative breast cancer (TNBC) cell line, HCC-1806, that was associated with a marked decrease in glutamine consumption, glutamate production, oxygen consumption, and the steady-state levels of glutathione and several tricarboxylic acid cycle intermediates. In contrast, no antiproliferative activity was observed in an estrogen receptor-positive cell line, T47D, and only modest effects on glutamine consumption and downstream metabolites were observed. Across a panel of breast cancer cell lines, GAC protein expression and glutaminase activity were elevated in the majority of TNBC cell lines relative to receptor positive cells. Furthermore, the TNBC subtype displayed the greatest sensitivity to CB-839 treatment and this sensitivity was correlated with (i) dependence on extracellular glutamine for growth, (ii) intracellular glutamate and glutamine levels, and (iii) GAC (but not KGA) expression, a potential biomarker for sensitivity. CB-839 displayed significant antitumor activity in two xenograft models: as a single agent in a patient-derived TNBC model and in a basal like HER2(+) cell line model, JIMT-1, both as a single agent and in combination with paclitaxel. Together, these data provide a strong rationale for the clinical investigation of CB-839 as a targeted therapeutic in patients with TNBC and other glutamine-dependent tumors.


Asunto(s)
Antineoplásicos/administración & dosificación , Bencenoacetamidas/farmacología , Inhibidores Enzimáticos/administración & dosificación , Glutaminasa/antagonistas & inhibidores , Neoplasias Basocelulares/tratamiento farmacológico , Tiadiazoles/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Administración Oral , Animales , Antineoplásicos/uso terapéutico , Bencenoacetamidas/uso terapéutico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Neoplasias Mamarias Experimentales , Ratones , Ratones SCID , Persona de Mediana Edad , Neoplasias Basocelulares/patología , Sulfuros/administración & dosificación , Sulfuros/uso terapéutico , Tiadiazoles/administración & dosificación , Tiadiazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Drug Metab Dispos ; 39(10): 1873-82, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21752943

RESUMEN

Carfilzomib [(2S)-N-[(S)-1-[(S)-4-methyl-1-[(R)-2-methyloxiran-2-yl]-1-oxopentan-2-ylcarbamoyl]-2-phenylethyl]-2-[(S)-2-(2-morpholinoacetamido)-4-phenylbutanamido]-4-methylpentanamide, also known as PR-171] is a selective, irreversible proteasome inhibitor that has shown encouraging results in clinical trials in multiple myeloma. In this study, the pharmacokinetics, pharmacodynamics, metabolism, distribution, and excretion of carfilzomib in Sprague-Dawley rats were characterized. After intravenous administration, the plasma concentration of carfilzomib declined rapidly in a biphasic manner. Carfilzomib displayed high plasma clearance [195-319 ml/(min · kg)], a short-terminal half-life (5-20 min), and rapid and wide tissue distribution in rats. The exposure to carfilzomib (C(max) and area under the curve) increased dose proportionally from 2 to 4 mg/kg but less than dose proportionally from 4 to 8 mg/kg. The high clearance was mediated predominantly by extrahepatic metabolism through peptidase cleavage and epoxide hydrolysis. Carfilzomib was excreted mainly as metabolites resulting from peptidase cleavage. Carfilzomib and its major metabolites in urine and bile accounted for approximately 26 and 31% of the total dose, respectively, for a total of 57% within 24 h postdose. Despite the high systemic clearance, potent proteasome inhibition was observed in blood and a variety of tissues. Together with rapid and irreversible target binding, the high clearance may provide an advantage in that "unnecessary" exposure to the drug is minimized and potential drug-related side effects may be reduced.


Asunto(s)
Oligopéptidos/metabolismo , Oligopéptidos/farmacocinética , Animales , Bilis/metabolismo , Compuestos Epoxi/metabolismo , Hidrólisis , Masculino , Oligopéptidos/farmacología , Péptido Hidrolasas/metabolismo , Ratas , Ratas Sprague-Dawley , Distribución Tisular
7.
Clin Cancer Res ; 17(9): 2734-43, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21364033

RESUMEN

PURPOSE: Bortezomib (Velcade), a dipeptide boronate 20S proteasome inhibitor and an approved treatment option for multiple myeloma, is associated with a treatment-emergent, painful peripheral neuropathy (PN) in more than 30% of patients. Carfilzomib, a tetrapeptide epoxyketone proteasome inhibitor, currently in clinical investigation in myeloma, is associated with low rates of PN. We sought to determine whether PN represents a target-mediated adverse drug reaction (ADR). EXPERIMENTAL DESIGN: Neurodegenerative effects of proteasome inhibitors were assessed in an in vitro model utilizing a differentiated neuronal cell line. Secondary targets of both inhibitors were identified by a multifaceted approach involving candidate screening, profiling with an activity-based probe, and database mining. Secondary target activity was measured in rats and patients receiving both inhibitors. RESULTS: Despite equivalent levels of proteasome inhibition, only bortezomib reduced neurite length, suggesting a nonproteasomal mechanism. In cell lysates, bortezomib, but not carfilzomib, significantly inhibited the serine proteases cathepsin G (CatG), cathepsin A, chymase, dipeptidyl peptidase II, and HtrA2/Omi at potencies near or equivalent to that for the proteasome. Inhibition of CatG was detected in splenocytes of rats receiving bortezomib and in peripheral blood mononuclear cells derived from bortezomib-treated patients. Levels of HtrA2/Omi, which is known to be involved in neuronal survival, were upregulated in neuronal cells exposed to both proteasome inhibitors but was inhibited only by bortezomib exposure. CONCLUSION: These data show that bortezomib-induced neurodegeneration in vitro occurs via a proteasome-independent mechanism and that bortezomib inhibits several nonproteasomal targets in vitro and in vivo, which may play a role in its clinical ADR profile.


Asunto(s)
Ácidos Borónicos/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Oligopéptidos/efectos adversos , Inhibidores de Proteasoma , Pirazinas/efectos adversos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Ácidos Borónicos/administración & dosificación , Bortezomib , Células Cultivadas , Cisteína Endopeptidasas/administración & dosificación , Cisteína Endopeptidasas/efectos adversos , Sistemas de Liberación de Medicamentos , Células Hep G2 , Humanos , Masculino , Modelos Biológicos , Oligopéptidos/administración & dosificación , Complejo de la Endopetidasa Proteasomal/metabolismo , Pirazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley
8.
PLoS One ; 6(12): e27996, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22216088

RESUMEN

Bortezomib (Velcade™) is a reversible proteasome inhibitor that is approved for the treatment of multiple myeloma (MM). Despite its demonstrated clinical success, some patients are deprived of treatment due to primary refractoriness or development of resistance during therapy. To investigate the role of the duration of proteasome inhibition in the anti-tumor response of bortezomib, we established clonal isolates of HT-29 adenocarcinoma cells adapted to continuous exposure of bortezomib. These cells were ~30-fold resistant to bortezomib. Two novel and distinct mutations in the ß5 subunit, Cys63Phe, located distal to the binding site in a helix critical for drug binding, and Arg24Cys, found in the propeptide region were found in all resistant clones. The latter mutation is a natural variant found to be elevated in frequency in patients with MM. Proteasome activity and levels of both the constitutive and immunoproteasome were increased in resistant cells, which correlated to an increase in subunit gene expression. These changes correlated with a more rapid recovery of proteasome activity following brief exposure to bortezomib. Increased recovery rate was not due to increased proteasome turnover as similar findings were seen in cells co-treated with cycloheximide. When we exposed resistant cells to the irreversible proteasome inhibitor carfilzomib we noted a slower rate of recovery of proteasome activity as compared to bortezomib in both parental and resistant cells. Importantly, carfilzomib maintained its cytotoxic potential in the bortezomib resistant cell lines. Therefore, resistance to bortezomib, can be overcome with irreversible inhibitors, suggesting prolonged proteasome inhibition induces a more potent anti-tumor response.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Ácidos Borónicos/uso terapéutico , Pirazinas/uso terapéutico , Adenocarcinoma/patología , Western Blotting , Bortezomib , Línea Celular Tumoral , Resistencia a Antineoplásicos , Ensayo de Inmunoadsorción Enzimática , Humanos
9.
Blood ; 114(16): 3439-47, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19671918

RESUMEN

Carfilzomib is a proteasome inhibitor in clinical development that primarily targets the chymotrypsin-like (CT-L) subunits in both the constitutive proteasome (c20S) and the immunoproteasome (i20S). To investigate the impact of inhibiting the CT-L activity with carfilzomib, we set out to quantitate the levels of CT-L subunits beta5 from the c20S and LMP7 from the i20S in normal and malignant hematopoietic cells. We found that the i20S is a major form of the proteasome expressed in cells of hematopoietic origin, including multiple myeloma (MM) CD138+ tumor cells. Although specific inhibition of either LMP7 or beta5 alone was insufficient to produce an antitumor response, inhibition of all proteasome subunits was cytotoxic to both hematologic tumor cells and peripheral blood mononuclear cells. However, selective inhibition of both beta5 and LMP7 was sufficient to induce an antitumor effect in MM, non-Hodgkin lymphoma, and leukemia cells while minimizing the toxicity toward nontransformed cells. In MM tumor cells, CT-L inhibition alone was sufficient to induce proapoptotic sequelae, including proteasome substrate accumulation, Noxa and caspase 3/7 induction, and phospho-eIF2alpha suppression. These data support a hypothesis that hematologic tumor cells are uniquely sensitive to CT-L inhibition and provide a mechanistic understanding of the clinical safety profile and antitumor activity of proteasome inhibitors.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Oligopéptidos/farmacología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Quimotripsina/antagonistas & inhibidores , Quimotripsina/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inducción Enzimática/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hematológicas/enzimología , Humanos , Oligopéptidos/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
10.
Nat Med ; 15(7): 781-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19525961

RESUMEN

The immunoproteasome, a distinct class of proteasome found predominantly in monocytes and lymphocytes, is known to shape the antigenic repertoire presented on class I major histocompatibility complexes (MHC-I). However, a specific role for the immunoproteasome in regulating other facets of immune responses has not been established. We describe here the characterization of PR-957, a selective inhibitor of low-molecular mass polypeptide-7 (LMP7, encoded by Psmb8), the chymotrypsin-like subunit of the immunoproteasome. PR-957 blocked presentation of LMP7-specific, MHC-I-restricted antigens in vitro and in vivo. Selective inhibition of LMP7 by PR-957 blocked production of interleukin-23 (IL-23) by activated monocytes and interferon-gamma and IL-2 by T cells. In mouse models of rheumatoid arthritis, PR-957 treatment reversed signs of disease and resulted in reductions in cellular infiltration, cytokine production and autoantibody levels. These studies reveal a unique role for LMP7 in controlling pathogenic immune responses and provide a therapeutic rationale for targeting LMP7 in autoimmune disorders.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Citocinas/biosíntesis , Complejos Multienzimáticos/antagonistas & inhibidores , Oligopéptidos/farmacología , Inhibidores de Proteasoma , Animales , Presentación de Antígeno/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Humanos , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Complejos Multienzimáticos/fisiología , Oligopéptidos/uso terapéutico , Complejo de la Endopetidasa Proteasomal
11.
J Med Chem ; 52(9): 3028-38, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19348473

RESUMEN

Proteasome inhibition has been validated as a therapeutic modality in the treatment of multiple myeloma and non-Hodgkin's lymphoma. Carfilzomib, an epoxyketone currently undergoing clinical trials in malignant diseases, is a highly selective inhibitor of the chymotrypsin-like (CT-L) activity of the proteasome. A chemistry effort was initiated to discover orally bioavailable analogues of carfilzomib, which would have potential for improved dosing flexibility and patient convenience over intravenously administered agents. The lead compound, 2-Me-5-thiazole-Ser(OMe)-Ser(OMe)-Phe-ketoepoxide (58) (PR-047), selectively inhibited CT-L activity of both the constitutive proteasome (beta5) and immunoproteasome (LMP7) and demonstrated an absolute bioavailability of up to 39% in rodents and dogs. It was well tolerated with repeated oral administration at doses resulting in >80% proteasome inhibition in most tissues and elicited an antitumor response equivalent to intravenously administered carfilzomib in multiple human tumor xenograft and mouse syngeneic models. The favorable pharmacologic profile supports its further development for the treatment of malignant diseases.


Asunto(s)
Dipéptidos/síntesis química , Dipéptidos/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Inhibidores de Proteasoma , Tiazoles/síntesis química , Tiazoles/farmacología , Administración Oral , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Disponibilidad Biológica , Línea Celular , Dipéptidos/química , Dipéptidos/farmacocinética , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Humanos , Cinética , Ratones , Oligopéptidos/química , Oligopéptidos/farmacocinética , Relación Estructura-Actividad , Especificidad por Sustrato , Tiazoles/química , Tiazoles/farmacocinética
12.
Curr Opin Drug Discov Devel ; 11(5): 616-25, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18729013

RESUMEN

The proteasome is a multicatalytic protease complex that mediates the controlled degradation of intracellular proteins, including key components of pathways that contribute to cancer cell growth and immune cell signaling. Validation for the proteasome as a therapeutic target in oncology was provided by bortezomib, a proteasome inhibitor that was approved for the treatment of multiple myeloma in 2003. Since that time, a number of structurally and mechanistically distinct proteasome inhibitors have entered clinical development in oncology. In this review, the chemical properties, preclinical antitumor activities and early clinical trials of these next-generation proteasome inhibitors are described and the potential for future proteasome inhibitor development in autoimmune indications is discussed.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Cisteína Proteinasa/uso terapéutico , Diseño de Fármacos , Factores Inmunológicos/uso terapéutico , Inhibidores de Proteasoma , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Enfermedades Autoinmunes/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/efectos adversos , Inhibidores de Cisteína Proteinasa/química , Evaluación Preclínica de Medicamentos , Humanos , Factores Inmunológicos/efectos adversos , Factores Inmunológicos/química , Estructura Molecular , Neoplasias/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Resultado del Tratamiento
13.
Cancer Res ; 67(13): 6383-91, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616698

RESUMEN

Clinical studies with bortezomib have validated the proteasome as a therapeutic target for the treatment of multiple myeloma and non-Hodgkin's lymphoma. However, significant toxicities have restricted the intensity of bortezomib dosing. Here we describe the antitumor activity of PR-171, a novel epoxyketone-based irreversible proteasome inhibitor that is currently in clinical development. In comparison to bortezomib, PR-171 exhibits equal potency but greater selectivity for the chymotrypsin-like activity of the proteasome. In cell culture, PR-171 is more cytotoxic than bortezomib following brief treatments that mimic the in vivo pharmacokinetics of both molecules. Hematologic tumor cells exhibit the greatest sensitivity to brief exposure, whereas solid tumor cells and nontransformed cell types are less sensitive to such treatments. Cellular consequences of PR-171 treatment include the accumulation of proteasome substrates and induction of cell cycle arrest and/or apoptosis. Administration of PR-171 to animals results in the dose-dependent inhibition of the chymotrypsin-like proteasome activity in all tissues examined with the exception of the brain. PR-171 is well tolerated when administered for either 2 or 5 consecutive days at doses resulting in >80% proteasome inhibition in blood and most tissues. In human tumor xenograft models, PR-171 mediates an antitumor response that is both dose and schedule dependent. The antitumor efficacy of PR-171 delivered on 2 consecutive days is stronger than that of bortezomib administered on its clinical dosing schedule. These studies show the tolerability, efficacy, and dosing flexibility of PR-171 and provide validation for the clinical testing of PR-171 in the treatment of hematologic malignancies using dose-intensive schedules.


Asunto(s)
Antineoplásicos/farmacología , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Apoptosis , Ácidos Borónicos/farmacología , Bortezomib , Quimotripsina/metabolismo , Quimotripsina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Masculino , Ratones , Trasplante de Neoplasias , Pirazinas/farmacología , Ratas , Ratas Sprague-Dawley
14.
Blood ; 110(9): 3281-90, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17591945

RESUMEN

The proteasome has emerged as an important target for cancer therapy with the approval of bortezomib, a first-in-class, reversible proteasome inhibitor, for relapsed/refractory multiple myeloma (MM). However, many patients have disease that does not respond to bortezomib, whereas others develop resistance, suggesting the need for other inhibitors with enhanced activity. We therefore evaluated a novel, irreversible, epoxomicin-related proteasome inhibitor, carfilzomib. In models of MM, this agent potently bound and specifically inhibited the chymotrypsin-like proteasome and immunoproteasome activities, resulting in accumulation of ubiquitinated substrates. Carfilzomib induced a dose- and time-dependent inhibition of proliferation, ultimately leading to apoptosis. Programmed cell death was associated with activation of c-Jun-N-terminal kinase, mitochondrial membrane depolarization, release of cytochrome c, and activation of both intrinsic and extrinsic caspase pathways. This agent also inhibited proliferation and activated apoptosis in patient-derived MM cells and neoplastic cells from patients with other hematologic malignancies. Importantly, carfilzomib showed increased efficacy compared with bortezomib and was active against bortezomib-resistant MM cell lines and samples from patients with clinical bortezomib resistance. Carfilzomib also overcame resistance to other conventional agents and acted synergistically with dexamethasone to enhance cell death. Taken together, these data provide a rationale for the clinical evaluation of carfilzomib in MM.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma , Ubiquitina , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Modelos Biológicos , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ubiquitina/antagonistas & inhibidores , Ubiquitina/metabolismo
15.
Surg Today ; 36(10): 934-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16998691

RESUMEN

A diagnosis of malignancy is reasonably assumed when a lesion is found at the hilum or bile ducts in a patient with jaundice who has never undergone biliary surgery. Although benign tumors occasionally develop in this location, preoperative recognition is difficult and most are treated as malignant lesions. We illustrate this clinical scenario in this case report of a granular cell tumor (GCT) that developed at the biliary bifurcation, necessitating right hemi-hepatectomy with extrahepatic biliary tree excision. We describe the clinical presentation, imaging findings, treatment, and histological findings of this tumor. Although rare, a GCT can develop at the hilum and mimic a malignant lesion such as cholangiocarcinoma (CC) radiologically. To our knowledge, this is the fourth report of a GCT at the hilum of the liver. However, the possibility of this tumor should be considered in the differential diagnosis of a lesion in this location.


Asunto(s)
Neoplasias de los Conductos Biliares/complicaciones , Colestasis Extrahepática/etiología , Tumor de Células Granulares/complicaciones , Conducto Hepático Común , Neoplasias de los Conductos Biliares/diagnóstico , Neoplasias de los Conductos Biliares/cirugía , Colangiografía , Colestasis Extrahepática/diagnóstico , Colestasis Extrahepática/cirugía , Diagnóstico Diferencial , Femenino , Tumor de Células Granulares/diagnóstico , Tumor de Células Granulares/cirugía , Hepatectomía , Humanos , Laparoscopía , Imagen por Resonancia Magnética , Persona de Mediana Edad
16.
J Immunol ; 174(9): 5288-97, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15843525

RESUMEN

TRAC-1 (T cell RING (really interesting new gene) protein identified in activation screen) is a novel E3 ubiquitin ligase identified from a retroviral vector-based T cell surface activation marker screen. The C-terminal truncated TRAC-1 specifically inhibited anti-TCR-mediated CD69 up-regulation in Jurkat cells, a human T leukemic cell line. In this study, we show that TRAC-1 is a RING finger ubiquitin E3 ligase with highest expression in lymphoid tissues. Point mutations that disrupt the Zn(2+)-chelating ability of its amino-terminal RING finger domain abolished TRAC-1's ligase activity and the dominant inhibitory effect of C-terminal truncated TRAC-1 on TCR stimulation. The results of in vitro biochemical studies indicate that TRAC-1 can stimulate the formation of both K48- and K63-linked polyubiquitin chains and therefore could potentially activate both degradative and regulatory ubiquitin-dependent pathways. Antisense oligonucleotides to TRAC-1 specifically reduced TRAC-1 mRNA levels in Jurkat and primary T cells and inhibited their activation in response to TCR cross-linking. Collectively, these results indicate that the E3 ubiquitin ligase TRAC-1 functions as a positive regulator of T cell activation.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Activación de Linfocitos/inmunología , Proteínas Nucleares/fisiología , Linfocitos T/enzimología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/fisiología , Regulación hacia Arriba/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia de Bases , Catálisis , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/aislamiento & purificación , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN , Humanos , Células Jurkat , Activación de Linfocitos/genética , Tejido Linfoide/citología , Tejido Linfoide/enzimología , Tejido Linfoide/inmunología , Datos de Secuencia Molecular , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Co-Represor 2 de Receptor Nuclear , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Poliubiquitina/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Proteínas Represoras , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/aislamiento & purificación , Regulación hacia Arriba/genética
18.
Drug Discov Today ; 8(16): 746-54, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12944097

RESUMEN

The ubiquitin system has been implicated in the pathogenesis of numerous disease states, including oncogenesis, inflammation, viral infection, CNS disorders and metabolic dysfunction. Ubiquitin conjugation and deconjugation to substrate proteins is carried out by multiple families of proteins, each with a defined role in the enzymatic cascade. This conjugation-deconjugation system parallels the kinase-phosphatase system in that both alter protein function by the addition and removal of post-translational modifiers. Our understanding of ubiquitin biology and strategies to interfere pharmacologically with the ubiquitin regulatory machinery is progressing rapidly. In light of increased interest in ubiquitin pathways as drug targets, we review the ubiquitin enzymatic cascades, highlighting therapeutic opportunities and enzymatic mechanisms. We also discuss the challenges of targeting this class of enzymes with small molecules, as well as current approaches and progress in drug discovery.


Asunto(s)
Ubiquitinas , Humanos , Ubiquitinas/genética , Ubiquitinas/metabolismo , Ubiquitinas/fisiología
19.
J Biol Chem ; 277(40): 37512-8, 2002 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-12167667

RESUMEN

Inteins are polypeptide sequences found in a small set of primarily bacterial proteins that promote the splicing of flanking pre-protein sequences to generate mature protein products. Inteins can be engineered in a "split and inverted" configuration such that the protein splicing product is a cyclic polypeptide consisting of the sequence linking two intein subdomains. We have engineered a split intein into a retroviral expression system to enable the intracellular delivery of a library of random cyclic peptides in human cells. Cyclization of peptides could be detected in cell lysates using mass spectrometry. A functional genetic screen to identify 5-amino acid-long cyclic peptides that block interleukin-4 mediated IgE class switching in B cells yielded 13 peptides that selectively inhibited germ line epsilon transcription. These results demonstrate the generation of cyclic peptide libraries in human cells and the power of functional screening to rapidly identify biologically active peptides.


Asunto(s)
Linfocitos B/inmunología , Proteínas Bacterianas , Interleucina-4/antagonistas & inhibidores , Biblioteca de Péptidos , Péptidos Cíclicos/química , Transducción de Señal/fisiología , Linfocitos B/efectos de los fármacos , Cianobacterias/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , AdnB Helicasas , Vectores Genéticos , Humanos , Empalme del ARN , Retroviridae , Transducción de Señal/efectos de los fármacos , Transfección
20.
IEEE Trans Inf Technol Biomed ; 6(1): 54-8, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11936597

RESUMEN

The aim of this study was to investigate the value of fractal dimension in separating normal and cancerous images, and to examine the relationship between fractal dimension and traditional texture analysis features. Forty-four normal images and 58 cancer images from sections of the colon were analyzed. A "leave-one-out" analysis approach was used to classify the samples into each group. With fractal analysis there was a highly significant difference between groups (p < 0.0001). Correlation and entropy features showed greater differences between the groups (p < 0.0001). Nevertheless, the addition of fractal analysis to the feature analysis improved the sensitivity from 90% to 95% and specificity from 86% to 93%.


Asunto(s)
Neoplasias del Colon/patología , Fractales , Estudios de Casos y Controles , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...