Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Nat Med ; 29(6): 1358-1363, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37248298

RESUMEN

D-2-hydroxyglutaric aciduria type II (D2HGA2) is a severe inborn disorder of metabolism caused by heterozygous R140 mutations in the IDH2 (isocitrate dehydrogenase 2) gene. Here we report the results of treatment of two children with D2HGA2, one of whom exhibited severe dilated cardiomyopathy, with the selective mutant IDH2 enzyme inhibitor enasidenib. In both children, enasidenib treatment led to normalization of D-2-hydroxyglutarate (D-2-HG) concentrations in body fluids. At doses of 50 mg and 60 mg per day, no side effects were observed, except for asymptomatic hyperbilirubinemia. For the child with cardiomyopathy, chronic D-2-HG inhibition was associated with improved cardiac function, and for both children, therapy was associated with improved daily functioning, global motility and social interactions. Treatment of the child with cardiomyopathy led to therapy-coordinated changes in serum phospholipid levels, which were partly recapitulated in cultured fibroblasts, associated with complex effects on lipid and redox-related gene pathways. These findings indicate that targeted inhibition of a mutant enzyme can partly reverse the pathology of a chronic neurometabolic genetic disorder.


Asunto(s)
Cardiomiopatías , Isocitrato Deshidrogenasa , Niño , Humanos , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/genética , Inhibidores Enzimáticos/efectos adversos , Células Germinativas/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Mutación/genética
2.
Blood ; 125(2): 296-303, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25398940

RESUMEN

Mutations of IDH1 and IDH2, which produce the oncometabolite 2-hydroxyglutarate (2HG), have been identified in several tumors, including acute myeloid leukemia. Recent studies have shown that expression of the IDH mutant enzymes results in high levels of 2HG and a block in cellular differentiation that can be reversed with IDH mutant-specific small-molecule inhibitors. To further understand the role of IDH mutations in cancer, we conducted mechanistic studies in the TF-1 IDH2 R140Q erythroleukemia model system and found that IDH2 mutant expression caused both histone and genomic DNA methylation changes that can be reversed when IDH2 mutant activity is inhibited. Specifically, histone hypermethylation is rapidly reversed within days, whereas reversal of DNA hypermethylation proceeds in a progressive manner over the course of weeks. We identified several gene signatures implicated in tumorigenesis of leukemia and lymphoma, indicating a selective modulation of relevant cancer genes by IDH mutations. As methylation of DNA and histones is closely linked to mRNA expression and differentiation, these results indicate that IDH2 mutant inhibition may function as a cancer therapy via histone and DNA demethylation at genes involved in differentiation and tumorigenesis.


Asunto(s)
Metilación de ADN/genética , Inhibidores Enzimáticos/farmacología , Histonas/genética , Isocitrato Deshidrogenasa/genética , Mutación , Transcriptoma/efectos de los fármacos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cromatografía Liquida , Histonas/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/genética , Compuestos de Fenilurea/farmacología , Análisis de Componente Principal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Espectrometría de Masas en Tándem
3.
Oncologist ; 19(6): 602-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24760710

RESUMEN

Mutations in the IDH1 and IDH2 (isocitrate dehydrogenase) genes have been discovered across a range of solid-organ and hematologic malignancies, including acute myeloid leukemia, glioma, chondrosarcoma, and cholangiocarcinoma. An intriguing aspect of IDH-mutant tumors is the aberrant production and accumulation of the oncometabolite 2-hydroxyglutarate (2-HG), which may play a pivotal oncogenic role in these malignancies. We describe the first reported case of an IDH1 p.R132L mutation in a patient with hormone receptor-positive (HR+) breast adenocarcinoma. This patient was initially treated for locally advanced disease, but then suffered a relapse and metastasis, at which point an IDH1-R132 mutation was discovered in an affected lymph node. The mutation was subsequently found in the primary tumor tissue and all metastatic sites, but not in an uninvolved lymph node. In addition, the patient's serum and urine displayed marked elevations in the concentration of 2-HG, significantly higher than that measured in six other patients with metastatic HR+ breast carcinoma whose tumors were found to harbor wild-type IDH1. In summary, IDH1 mutations may impact a rare subgroup of patients with breast adenocarcinoma. This may suggest future avenues for disease monitoring through noninvasive measurement of 2-HG, as well as for the development and study of targeted therapies against the aberrant IDH1 enzyme.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Mama/genética , Isocitrato Deshidrogenasa/genética , Neoplasias Hormono-Dependientes/genética , Adenocarcinoma/sangre , Adenocarcinoma/patología , Adenocarcinoma/orina , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Neoplasias de la Mama/orina , Femenino , Glutaratos/sangre , Glutaratos/orina , Humanos , Persona de Mediana Edad , Mutación , Neoplasias Hormono-Dependientes/sangre , Neoplasias Hormono-Dependientes/patología , Neoplasias Hormono-Dependientes/orina
4.
Clin Cancer Res ; 20(7): 1884-90, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24478380

RESUMEN

PURPOSE: Mutations in the IDH1 and IDH2 (IDH1/2) genes occur in approximately 20% of intrahepatic cholangiocarcinoma and lead to accumulation of 2-hydroxyglutarate (2HG) in the tumor tissue. However, it remains unknown whether IDH1/2 mutations can lead to high levels of 2HG circulating in the blood and whether serum 2HG can be used as a biomarker for IDH1/2 mutational status and tumor burden in intrahepatic cholangiocarcinoma. EXPERIMENTAL DESIGN: We initially measured serum 2HG concentration in blood samples collected from 31 patients with intrahepatic cholangiocarcinoma in a screening cohort. Findings were validated across 38 resected patients with intrahepatic cholangiocarcinoma from a second cohort with tumor volume measures. Circulating levels of 2HG were evaluated relative to IDH1/2 mutational status, tumor burden, and a number of clinical variables. RESULTS: Circulating levels of 2HG in the screening cohort were significantly elevated in patients with IDH1/2-mutant (median, 478 ng/mL) versus IDH1/2-wild-type (median, 118 ng/mL) tumors (P < 0.001). This significance was maintained in the validation cohort (343 ng/mL vs. 55 ng/mL, P < 0.0001) and levels of 2HG directly correlated with tumor burden in IDH1/2-mutant cases (P < 0.05). Serum 2HG levels ≥170 ng/mL could predict the presence of an IDH1/2 mutation with a sensitivity of 83% and a specificity of 90%. No differences were noted between the allelic variants IDH1 or IDH2 in regard to the levels of circulating 2HG. CONCLUSIONS: This study indicates that circulating 2HG may be a surrogate biomarker of IDH1 or IDH2 mutation status in intrahepatic cholangiocarcinoma and that circulating 2HG levels may correlate directly with tumor burden. Clin Cancer Res; 20(7); 1884-90. ©2014 AACR.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Biomarcadores de Tumor/sangre , Colangiocarcinoma/genética , Glutaratos/sangre , Isocitrato Deshidrogenasa/genética , Adulto , Anciano , Neoplasias de los Conductos Biliares/sangre , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/sangre , Colangiocarcinoma/patología , Femenino , Humanos , Isocitrato Deshidrogenasa/sangre , Masculino , Persona de Mediana Edad , Mutación
5.
Nat Rev Drug Discov ; 12(10): 743-55, 2013 10.
Artículo en Inglés | MEDLINE | ID: mdl-24008432

RESUMEN

As diagnostic tests become increasingly important for optimizing the use of drugs to treat cancers, the co-development of a targeted therapy and its companion diagnostic test is becoming more prevalent and necessary. In July 2011, the US Food and Drug Administration released a draft guidance that gave the agency's formal definition of companion diagnostics and introduced a drug-diagnostic co-development process for gaining regulatory approval. Here, we identify areas of drug-diagnostic co-development that were either not covered by the guidance or that would benefit from increased granularity, including how to determine when clinical studies should be limited to biomarker-positive patients, defining the diagnostically selected patient population in which to use a companion diagnostic, and defining and clinically validating a biomarker signature for assays that use more than one biomarker. We propose potential approaches that sponsors could use to deal with these challenges and provide strategies to help guide the future co-development of drugs and diagnostics.


Asunto(s)
Antineoplásicos/farmacología , Técnicas de Diagnóstico Molecular , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Biomarcadores/análisis , Biomarcadores/metabolismo , Aprobación de Recursos , Aprobación de Drogas , Diseño de Fármacos , Humanos , Neoplasias/diagnóstico , Neoplasias/patología , Estados Unidos , United States Food and Drug Administration
6.
Clin Cancer Res ; 19(11): 2804-9, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23553847

RESUMEN

For a new therapy to qualify for the accelerated approval pathway, it must treat a serious disease for which there is "unmet medical need"--defined as providing a therapy where none exists or providing a therapy that may be potentially superior to existing therapy. The increasing number of available therapies, coupled with the lack of accepted endpoints considered "reasonably likely to predict clinical benefit" and the lack of clarity early in development about circumstances in which a new product will qualify for accelerated approval, is pushing developers to pursue accelerated approval in heavily pretreated patients to fulfill an unmet need. To optimize the accelerated approval pathway, we propose here a reevaluation of what constitutes "unmet medical need" and "available therapy" in oncology. We also discuss ways for new endpoints to become qualified for use in supporting accelerated approval, and propose a structured process for pursuing accelerated approval.


Asunto(s)
Antineoplásicos , Aprobación de Drogas , Humanos , Estados Unidos , United States Food and Drug Administration
7.
Science ; 340(6132): 622-6, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23558173

RESUMEN

A number of human cancers harbor somatic point mutations in the genes encoding isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2). These mutations alter residues in the enzyme active sites and confer a gain-of-function in cancer cells, resulting in the accumulation and secretion of the oncometabolite (R)-2-hydroxyglutarate (2HG). We developed a small molecule, AGI-6780, that potently and selectively inhibits the tumor-associated mutant IDH2/R140Q. A crystal structure of AGI-6780 complexed with IDH2/R140Q revealed that the inhibitor binds in an allosteric manner at the dimer interface. The results of steady-state enzymology analysis were consistent with allostery and slow-tight binding by AGI-6780. Treatment with AGI-6780 induced differentiation of TF-1 erythroleukemia and primary human acute myelogenous leukemia cells in vitro. These data provide proof-of-concept that inhibitors targeting mutant IDH2/R140Q could have potential applications as a differentiation therapy for cancer.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hematopoyesis/efectos de los fármacos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/enzimología , Compuestos de Fenilurea/farmacología , Sulfonamidas/farmacología , Sitio Alostérico , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Eritropoyesis/efectos de los fármacos , Regulación Leucémica de la Expresión Génica , Glutaratos/metabolismo , Humanos , Isocitrato Deshidrogenasa/química , Isocitrato Deshidrogenasa/metabolismo , Leucemia Eritroblástica Aguda , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Terapia Molecular Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Compuestos de Fenilurea/química , Compuestos de Fenilurea/metabolismo , Mutación Puntual , Multimerización de Proteína , Estructura Secundaria de Proteína , Bibliotecas de Moléculas Pequeñas , Sulfonamidas/química , Sulfonamidas/metabolismo
9.
Blood ; 120(23): 4649-52, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23074281

RESUMEN

Mutations of genes encoding isocitrate dehydrogenase (IDH1 and IDH2) have been recently described in acute myeloid leukemia (AML). Serum and myeloblast samples from patients with IDH-mutant AML contain high levels of the metabolite 2-hydroxyglutarate (2-HG), a product of the altered IDH protein. In this prospective study, we sought to determine whether 2-HG can potentially serve as a noninvasive biomarker of disease burden through serial measurements in patients receiving conventional therapy for newly diagnosed AML. Our data demonstrate that serum, urine, marrow aspirate, and myeloblast 2-HG levels are significantly higher in IDH-mutant patients, with a correlation between baseline serum and urine 2-HG levels. Serum and urine 2-HG, along with IDH1/2-mutant allele burden in marrow, decreased with response to treatment. 2-HG decrease was more rapid with induction chemotherapy compared with DNA-methyltransferase inhibitor therapy. Our data suggest that serum or urine 2-HG may serve as noninvasive biomarkers of disease activity for IDH-mutant AML.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Glutaratos/metabolismo , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/metabolismo , Enfermedad Aguda , Anciano , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Citarabina/administración & dosificación , Análisis Mutacional de ADN , Decitabina , Femenino , Glutaratos/sangre , Glutaratos/orina , Células Precursoras de Granulocitos/metabolismo , Humanos , Idarrubicina/administración & dosificación , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Leucemia Mieloide/genética , Masculino , Persona de Mediana Edad , Mutación , Estudios Prospectivos , Factores de Tiempo
11.
Oncologist ; 17(1): 72-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22180306

RESUMEN

Cancers of origin in the gallbladder and bile ducts are rarely curable with current modalities of cancer treatment. Our clinical application of broad-based mutational profiling for patients diagnosed with a gastrointestinal malignancy has led to the novel discovery of mutations in the gene encoding isocitrate dehydrogenase 1 (IDH1) in tumors from a subset of patients with cholangiocarcinoma. A total of 287 tumors from gastrointestinal cancer patients (biliary tract, colorectal, gastroesophageal, liver, pancreatic, and small intestine carcinoma) were tested during routine clinical evaluation for 130 site-specific mutations within 15 cancer genes. Mutations were identified within a number of genes, including KRAS (35%), TP53 (22%), PIK3CA (10%), BRAF (7%), APC (6%), NRAS (3%), AKT1 (1%), CTNNB1 (1%), and PTEN (1%). Although mutations in the metabolic enzyme IDH1 were rare in the other common gastrointestinal malignancies in this series (2%), they were found in three tumors (25%) of an initial series of 12 biliary tract carcinomas. To better define IDH1 and IDH2 mutational status, an additional 75 gallbladder and bile duct cancers were examined. Combining these cohorts of biliary cancers, mutations in IDH1 and IDH2 were found only in cholangiocarcinomas of intrahepatic origin (nine of 40, 23%) and in none of the 22 extrahepatic cholangiocarcinomas and none of the 25 gallbladder carcinomas. In an analysis of frozen tissue specimens, IDH1 mutation was associated with highly elevated tissue levels of the enzymatic product 2-hydroxyglutarate. Thus, IDH1 mutation is a molecular feature of cholangiocarcinomas of intrahepatic origin. These findings define a specific metabolic abnormality in this largely incurable type of gastrointestinal cancer and present a potentially new target for therapy.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/genética , Neoplasias de la Vesícula Biliar/genética , Isocitrato Deshidrogenasa/genética , Mutación , Adolescente , Adulto , Neoplasias de los Conductos Biliares/enzimología , Niño , Colangiocarcinoma/enzimología , Femenino , Neoplasias de la Vesícula Biliar/enzimología , Genotipo , Humanos , Isocitrato Deshidrogenasa/metabolismo , Masculino , Persona de Mediana Edad , Adulto Joven
12.
J Neurooncol ; 107(1): 197-205, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22015945

RESUMEN

Mutations of the isocitrate dehydrogenase 1 and 2 genes (IDH1 and IDH2) are commonly found in primary brain cancers. We previously reported that a novel enzymatic activity of these mutations results in the production of the putative oncometabolite, R(-)-2-hydroxyglutarate (2-HG). Here we investigated the ability of magnetic resonance spectroscopy (MRS) to detect 2-HG production in order to non-invasively identify patients with IDH1 mutant brain tumors. Patients with intrinsic glial brain tumors (n = 27) underwent structural and spectroscopic magnetic resonance imaging prior to surgery. 2-HG levels from MRS data were quantified using LC-Model software, based upon a simulated spectrum obtained from a GAMMA library added to the existing prior knowledge database. The resected tumors were then analyzed for IDH1 mutational status by genomic DNA sequencing, Ki-67 proliferation index by immunohistochemistry, and concentrations of 2-HG and other metabolites by liquid chromatography-mass spectrometry (LC-MS). MRS detected elevated 2-HG levels in gliomas with IDH1 mutations compared to those with wild-type IDH1 (P = 0.003). The 2-HG levels measured in vivo with MRS were significantly correlated with those measured ex vivo from the corresponding tumor samples using LC-MS (r (2) = 0.56; P = 0.0001). Compared with wild-type tumors, those with IDH1 mutations had elevated choline (P = 0.01) and decreased glutathione (P = 0.03) on MRS. Among the IDH1 mutated gliomas, quantitative 2-HG values were correlated with the Ki-67 proliferation index of the tumors (r ( 2 ) = 0.59; P = 0.026). In conclusion, water-suppressed proton ((1)H) MRS provides a non-invasive measure of 2-HG in gliomas, and may serve as a potential biomarker for patients with IDH1 mutant brain tumors. In addition to 2-HG, alterations in several other metabolites measured by MRS correlate with IDH1 mutation status.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Glioma/genética , Glioma/metabolismo , Glutaratos/metabolismo , Isocitrato Deshidrogenasa/genética , Espectroscopía de Resonancia Magnética , Mutación/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Cromatografía Liquida , ADN de Neoplasias/genética , Femenino , Estudios de Seguimiento , Glioma/patología , Humanos , Imagen por Resonancia Magnética , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Pronóstico , Estudios Prospectivos , Adulto Joven
13.
J Exp Med ; 207(2): 339-44, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20142433

RESUMEN

Mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2), are present in most gliomas and secondary glioblastomas, but are rare in other neoplasms. IDH1/2 mutations are heterozygous, and affect a single arginine residue. Recently, IDH1 mutations were identified in 8% of acute myelogenous leukemia (AML) patients. A glioma study revealed that IDH1 mutations cause a gain-of-function, resulting in the production and accumulation of 2-hydroxyglutarate (2-HG). Genotyping of 145 AML biopsies identified 11 IDH1 R132 mutant samples. Liquid chromatography-mass spectrometry metabolite screening revealed increased 2-HG levels in IDH1 R132 mutant cells and sera, and uncovered two IDH2 R172K mutations. IDH1/2 mutations were associated with normal karyotypes. Recombinant IDH1 R132C and IDH2 R172K proteins catalyze the novel nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reduction of alpha-ketoglutarate (alpha-KG) to 2-HG. The IDH1 R132C mutation commonly found in AML reduces the affinity for isocitrate, and increases the affinity for NADPH and alpha-KG. This prevents the oxidative decarboxylation of isocitrate to alpha-KG, and facilitates the conversion of alpha-KG to 2-HG. IDH1/2 mutations confer an enzymatic gain of function that dramatically increases 2-HG in AML. This provides an explanation for the heterozygous acquisition of these mutations during tumorigenesis. 2-HG is a tractable metabolic biomarker of mutant IDH1/2 enzyme activity.


Asunto(s)
Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/enzimología , Mutación , Catálisis , Glutaratos/sangre , Humanos , Isocitrato Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/genética , NADP/metabolismo , Proteínas Recombinantes/metabolismo
14.
Clin Cancer Res ; 14(5): 1446-54, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18316568

RESUMEN

PURPOSE: Proteasome inhibition results in cytotoxicity to the leukemia stem cell in vitro. We conducted this phase I study to determine if the proteasome inhibitor bortezomib could be safely added to induction chemotherapy in patients with acute myelogenous leukemia (AML). EXPERIMENTAL DESIGN: Bortezomib was given on days 1, 4, 8, and 11 at doses of 0.7, 1.0, 1.3, or 1.5 mg/m(2) with idarubicin 12 mg/m(2) on days 1 to 3 and cytarabine 100 mg/m(2)/day on days 1 to 7. RESULTS: A total of 31 patients were enrolled. The median age was 62 years, and 16 patients were male. Nine patients had relapsed AML (ages, 18-59 years, n = 4 and > or = 60 years, n = 5). There were 22 patients of > or = 60 years with previously untreated AML (eight with prior myelodysplasia/myeloproliferative disorder or cytotoxic therapy). All doses of bortezomib, up to and including 1.5 mg/m(2), were tolerable. Nonhematologic grade 3 or greater toxicities included 12 hypoxia (38%; 11 were grade 3), 4 hyperbilirubinemia (13%), and 6 elevated aspartate aminotransferase (19%). Overall, 19 patients (61%) achieved complete remission (CR) and three had CR with incomplete platelet recovery. Pharmacokinetic studies revealed that the total body clearance of bortezomib decreased significantly (P < 0.01, N = 26) between the first (mean +/- SD, 41.9 +/- 17.1 L/h/m(2)) and third (18.4 +/- 7.0 L/h/m(2)) doses. Increased bone marrow expression of CD74 was associated with CR. CONCLUSIONS: The combination of bortezomib, idarubicin, and cytarabine showed a good safety profile. The recommended dose of bortezomib for phase II studies with idarubicin and cytarabine is 1.5 mg/m(2).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Leucemia Mieloide Aguda/metabolismo , Adolescente , Adulto , Anciano , Biomarcadores de Tumor , Ácidos Borónicos/administración & dosificación , Bortezomib , Estudios de Cohortes , Citarabina/administración & dosificación , Femenino , Perfilación de la Expresión Génica , Humanos , Idarrubicina/administración & dosificación , Leucemia Mieloide Aguda/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Análisis de Secuencia por Matrices de Oligonucleótidos , Pirazinas/administración & dosificación , Distribución Tisular , Resultado del Tratamiento
15.
J Thorac Oncol ; 3(1): 68-74, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18166843

RESUMEN

INTRODUCTION: Bortezomib is a small-molecule proteasome inhibitor with single-agent activity in patients with non-small cell lung carcinoma (NSCLC) and synergy with gemcitabine in preclinical studies. The combination of gemcitabine and carboplatin is an accepted first-line treatment for advanced NSCLC. We conducted a phase I study of gemcitabine and carboplatin in combination with bortezomib. METHODS: Bortezomib was administered on days 1, 4, 8, and 11, after gemcitabine on days 1 and 8, and carboplatin on day 1 of a 21-day cycle. Three escalating dose levels were evaluated: bortezomib 1.0 mg/m2/gemcitabine 800 mg/m2, bortezomib 1.0 mg/m2/gemcitabine 1000 mg/m2, and bortezomib 1.3 mg/m2/gemcitabine 1000 mg/m2, in combination with carboplatin AUC 5.0. RESULTS: Twenty-six patients with advanced NSCLC were treated; 21 were chemotherapy-naive. The median age was 59 years (range, 34-74), and 23 patients were stage IV. The Karnofsky performance score was 80% in 16 patients. Dose-limiting toxicities were grade 3 thrombocytopenia with bleeding and febrile neutropenia accompanied by grade 4 thrombocytopenia and grade 3 hyponatremia. The maximum-tolerated dose was defined as bortezomib 1.0 mg/m2, gemcitabine 1000 mg/m2, and carboplatin AUC 5.0. The most common grade 3/4 toxicities were thrombocytopenia (rarely associated with bleeding), and neutropenia. Nine of 26 patients (35%) achieved partial response, and eight patients had stable disease. CONCLUSIONS: The combination of bortezomib 1.0 mg/m2, gemcitabine 1000 mg/m2, and carboplatin AUC 5.0 demonstrated manageable toxicities and encouraging activity in NSCLC. This regimen was used in a phase II study.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Inhibidores de Proteasoma , Adulto , Anciano , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/toxicidad , Antineoplásicos/administración & dosificación , Antineoplásicos/toxicidad , Ácidos Borónicos/administración & dosificación , Bortezomib , California , Carboplatino/administración & dosificación , Carboplatino/toxicidad , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Desoxicitidina/toxicidad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Neutropenia/inducido químicamente , Pirazinas/administración & dosificación , Trombocitopenia/inducido químicamente , Resultado del Tratamiento , Gemcitabina
16.
Blood ; 110(10): 3557-60, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17690257

RESUMEN

Initial analysis of the Assessment of Proteasome Inhibition for Extending Remissions (APEX) trial of relapsed multiple myeloma patients showed significantly longer time to progression, higher response rate, and improved survival with single-agent bortezomib versus high-dose dexamethasone. In this updated analysis (median follow-up: 22 months), survival was assessed in both arms, and efficacy updated for the bortezomib arm. Median survival was 29.8 months for bortezomib versus 23.7 months for dexamethasone, a 6-month benefit, despite substantial crossover from dexamethasone to bortezomib. Overall and complete response rates with bortezomib were 43% and 9%, respectively; among responding patients, 56% improved response with longer therapy beyond initial response, leading to continued improvement in overall quality of response. Higher response quality (100% M-protein reduction) was associated with longer response duration; response duration was not associated with time to response. These data confirm the activity of bortezomib and support extended treatment in relapsed multiple myeloma patients tolerating therapy.


Asunto(s)
Ácidos Borónicos/uso terapéutico , Dexametasona/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/uso terapéutico , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Ácidos Borónicos/efectos adversos , Bortezomib , Supervivencia sin Enfermedad , Estudios de Seguimiento , Humanos , Mieloma Múltiple/mortalidad , Pirazinas/efectos adversos , Recurrencia , Factores de Tiempo , Resultado del Tratamiento
17.
Cancer Res ; 67(6): 2783-90, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363600

RESUMEN

Bortezomib is a proteasome inhibitor with proven efficacy in multiple myeloma and non-Hodgkin's lymphoma. This study reports the effects of bortezomib in B-cell lymphoma cell lines with differing sensitivity to bortezomib to investigate factors that influence sensitivity. Bortezomib induced a time- and concentration-dependent reduction in cell viability in five lymphoma cell lines, with EC(50) values ranging from 6 nmol/L (DHL-7 cells) to 25 nmol/L (DHL-4 cells) after 72 h. Bortezomib cytotoxicity was independent of p53 function, as all cell lines exhibited mutations by sequence analysis. The difference in sensitivity was not explained by proteasome or nuclear factor-kappaB (NF-kappaB) inhibition as these were similar in the most and least sensitive cells. NF-kappaB inhibition was less marked than that of a specific NF-kappaB inhibitor, Bay 11-7082. Cell cycle analysis showed a marked G(2)-arrested population in the least sensitive DHL-4 line only, an effect that was not present with Bay 11-7082 treatment. Conversely, in DHL-7 cells, bortezomib treatment resulted in cells moving into an aberrant mitosis, indicative of mitotic catastrophe that may contribute to increased sensitivity to bortezomib. These studies show that although bortezomib treatment had similar effects on apoptotic and NF-kappaB signaling pathways in these cell lines, different cell cycle effects were observed and induction of a further mechanism of cell death, mitotic catastrophe, was observed in the more sensitive cell line, which may provide some pointers to the difference in sensitivity between cell lines. An improved understanding of how DHL-7 cells abrogate the G(2)-M cell cycle checkpoint may help identify targets to increase the efficacy of bortezomib.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Linfoma de Células B/tratamiento farmacológico , Mitosis/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Pirazinas/farmacología , Proteína p53 Supresora de Tumor/fisiología , Bortezomib , Caspasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Activación Enzimática , Humanos , Linfoma de Células B/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonas/farmacología
18.
Blood ; 109(8): 3177-88, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17185464

RESUMEN

The aims of this study were to assess the feasibility of prospective pharmacogenomics research in multicenter international clinical trials of bortezomib in multiple myeloma and to develop predictive classifiers of response and survival with bortezomib. Patients with relapsed myeloma enrolled in phase 2 and phase 3 clinical trials of bortezomib and consented to genomic analyses of pretreatment tumor samples. Bone marrow aspirates were subject to a negative-selection procedure to enrich for tumor cells, and these samples were used for gene expression profiling using DNA microarrays. Data quality and correlations with trial outcomes were assessed by multiple groups. Gene expression in this dataset was consistent with data published from a single-center study of newly diagnosed multiple myeloma. Response and survival classifiers were developed and shown to be significantly associated with outcome via testing on independent data. The survival classifier improved on the risk stratification provided by the International Staging System. Predictive models and biologic correlates of response show some specificity for bortezomib rather than dexamethasone. Informative gene expression data and genomic classifiers that predict clinical outcome can be derived from prospective clinical trials of new anticancer agents.


Asunto(s)
Ácidos Borónicos/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Modelos Biológicos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Inhibidores de Proteasas/administración & dosificación , Pirazinas/administración & dosificación , Antineoplásicos Hormonales/administración & dosificación , Bortezomib , Dexametasona/administración & dosificación , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Farmacogenética , Valor Predictivo de las Pruebas , Estudios Prospectivos , Inhibidores de Proteasoma , Recurrencia , Resultado del Tratamiento
19.
Clin Cancer Res ; 12(19): 5887-94, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17020997

RESUMEN

PURPOSE: The purpose of this study is to delineate the biological significance of IkappaB kinase (IKK) beta inhibition in multiple myeloma cells in the context of bone marrow stromal cells (BMSC) using a novel IKKbeta inhibitor MLN120B. EXPERIMENTAL DESIGN: Growth-inhibitory effect of MLN120B in multiple myeloma cells in the presence of cytokines [interleukin-6 (IL-6) and insulin-like growth factor-I (IGF-1)], conventional agents (dexamethasone, melphalan, and doxorubicin), or BMSC was assessed in vitro. In vivo anti-multiple myeloma activity of MLN120B was evaluated in severe combined immunodeficient (SCID)-hu model. RESULTS: MLN120B inhibits both baseline and tumor necrosis factor-alpha-induced nuclear factor-kappaB activation, associated with down-regulation of IkappaBalpha and p65 nuclear factor-kappaB phosphorylation. MLN120B triggers 25% to 90% growth inhibition in a dose-dependent fashion in multiple myeloma cell lines and significantly augments tumor necrosis factor-alpha-induced cytotoxicity in MM.1S cells. MLN120B augments growth inhibition triggered by doxorubicin and melphalan in both RPMI 8226 and IL-6-dependent INA6 cell lines. Neither IL-6 nor IGF-1 overcomes the growth-inhibitory effect of MLN120B. MLN120B inhibits constitutive IL-6 secretion by BMSCs by 70% to 80% without affecting viability. Importantly, MLN120B almost completely blocks stimulation of MM.1S, U266, and INA6 cell growth, as well as IL-6 secretion from BMSCs, induced by multiple myeloma cell adherence to BMSCs. MLN120B overcomes the protective effect of BMSCs against conventional (dexamethasone) therapy. CONCLUSIONS: Our data show that the novel IKKbeta inhibitor MLN120B induces growth inhibition of multiple myeloma cells in SCID-hu mouse model. These studies provide the framework for clinical evaluation of MLN120B, alone and in combined therapies, trials of these novel agents to improve patient outcome in multiple myeloma.


Asunto(s)
Apoptosis/efectos de los fármacos , Médula Ósea/patología , Proliferación Celular/efectos de los fármacos , Quinasa I-kappa B/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Inhibidores Enzimáticos , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Interleucina-6/farmacología , Ratones , Ratones SCID , Mieloma Múltiple/patología , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Br J Haematol ; 134(4): 391-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16882131

RESUMEN

Bortezomib is the first proteasome inhibitor to be approved for use in haematological malignancies. Although a rash has been described as a common adverse event associated with the drug, it has not been well characterised. Based on three phase II studies of bortezomib in patients with non-Hodgkin lymphoma (140 assessable patients), we identified 26 patients who developed a unique erythematous maculopapular rash during treatment, six of whom underwent cutaneous biopsy. Punch biopsy in six patients revealed a perivascular lymphocytic infiltrate without evidence of lymphoma, consistent with a non-necrotising cutaneous vasculitis. The combined overall response rate was 41%. The response in the 26 patients who developed a rash was 73%, compared with 33% in patients who did not. The odds ratio for response given the development of a rash was 4.6 (95% CI, 1.7-12.4, P = 0.001). This is the first report to characterise a vasculitic rash associated with bortezomib, and to show a relationship between development of the rash and response to treatment. Unlike classic hypersensitivity type reactions, this vasculitic rash may not necessarily prompt cessation of drug. In fact, the development of an isolated cutaneous vasculitis may portend a better clinical response to bortezomib in some patients.


Asunto(s)
Ácidos Borónicos/efectos adversos , Hipersensibilidad a las Drogas , Linfoma no Hodgkin/tratamiento farmacológico , Inhibidores de Proteasas/efectos adversos , Pirazinas/efectos adversos , Enfermedades Cutáneas Vasculares/inducido químicamente , Anciano , Ácidos Borónicos/uso terapéutico , Bortezomib , Hipersensibilidad a las Drogas/patología , Femenino , Humanos , Modelos Logísticos , Linfoma no Hodgkin/patología , Masculino , Persona de Mediana Edad , Pronóstico , Inhibidores de Proteasas/uso terapéutico , Pirazinas/uso terapéutico , Piel/patología , Enfermedades Cutáneas Vasculares/patología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...