Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Clin Cancer Res ; 27(15): 4142-4146, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33712511

RESUMEN

On April 10, 2020, the FDA approved selumetinib (KOSELUGO, AstraZeneca) for the treatment of pediatric patients 2 years of age and older with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas. Approval was based on demonstration of a durable overall response rate per Response Evaluation in Neurofibromatosis and Schwannomatosis criteria and supported by observed clinical improvements in plexiform neurofibroma-related symptoms and functional impairments in 50 pediatric patients with inoperable plexiform neurofibromas in a single-arm, multicenter trial. The overall reponse rate per NCI investigator assessment was 66% (95% confidence interval, 51-79) with at least 12 months of follow-up. The median duration of response was not reached, and 82% of responding patients experienced duration of response ≥12 months. Clinical outcome assessment endpoints provided supportive efficacy data. Risks of selumetinib are consistent with MAPK (MEK) inhibitor class effects, including ocular, cardiac, musculoskeletal, gastrointestinal, and dermatologic toxicities. Safety was assessed across a pooled database of 74 pediatric patients with plexiform neurofibromas and supported by adult and pediatric selumetinib clinical trial data in cancer indications. The benefit-risk assessment for selumetinib in patients with inoperable plexiform neurofibromas was considered favorable.


Asunto(s)
Bencimidazoles/uso terapéutico , Aprobación de Drogas , Neurofibroma Plexiforme/tratamiento farmacológico , Adolescente , Niño , Preescolar , Femenino , Humanos , Masculino , Estados Unidos
2.
Clin Cancer Res ; 27(4): 928-932, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-32967940

RESUMEN

The FDA-approved entrectinib on August 15, 2019, for the treatment of adult and pediatric patients 12 years of age and older with solid tumors that have a neurotrophic tyrosine receptor kinase (NTRK) gene fusion without a known acquired resistance mutation, are metastatic or where surgical resection is likely to result in severe morbidity, and have progressed following treatment or have no satisfactory alternative therapy. Approval was based on demonstration of a durable overall response rate of 57% (95% confidence interval: 43-71), including a complete response rate of 7%, among 54 entrectinib-treated patients with 10 different tumor types harboring an NTRK fusion enrolled in one of three single-arm clinical trials. The durations of response ranged from 2.8 months to 26.0+ months; 68% of responses lasted ≥ 6 months. The most serious toxicities of entrectinib are congestive heart failure, central nervous system effects, skeletal fractures, hepatotoxicity, hyperuricemia, QT prolongation, and vision disorders. Adverse reactions were manageable through dose interruptions (46%), dose reductions (29%), or discontinuation of entrectinib (9%). This is the third approval of a cancer drug for treatment of a tissue agnostic, biomarker-defined cancer.


Asunto(s)
Benzamidas/administración & dosificación , Aprobación de Drogas , Indazoles/administración & dosificación , Neoplasias/tratamiento farmacológico , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/administración & dosificación , Benzamidas/efectos adversos , Humanos , Indazoles/efectos adversos , Neoplasias/genética , Proteínas de Fusión Oncogénica/genética , Inhibidores de Proteínas Quinasas/efectos adversos , Receptor trkA/antagonistas & inhibidores , Receptor trkA/genética , Receptor trkB/antagonistas & inhibidores , Receptor trkB/genética , Receptor trkC/antagonistas & inhibidores , Receptor trkC/genética , Estados Unidos , United States Food and Drug Administration/legislación & jurisprudencia
3.
Clin Cancer Res ; 23(12): 2924-2927, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28213365

RESUMEN

The FDA approved TAS-102 (Lonsurf; Taiho Oncology, Inc.) for the treatment of patients with metastatic colorectal cancer (mCRC) who have been previously treated with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy; an anti-VEGF biological therapy; and if RAS wild type, an anti-EGFR therapy. In an international, multicenter, double-blinded, placebo-controlled trial (TPU-TAS-102-301, herein referred to as RECOURSE), 800 patients with previously treated mCRC were randomly allocated (2:1) to receive either TAS-102 35 mg/m2 orally twice daily after meals on days 1 through 5 and 8 through 12 of each 28-day cycle (n = 534) or matching placebo (n = 266). The trial demonstrated a statistically significant improvement in overall survival for those randomized to receive TAS-102, with a median survival of 7.1 months in the TAS-102 arm [confidence interval (CI), 6.5-7.8] and 5.3 months in the placebo arm [CI, 4.6-6.0; hazard ratio (HR), 0.68; 95% CI, 0.58-0.81; P < 0.001, stratified log-rank test]. The trial also demonstrated a statistically significant prolongation of progression-free survival (HR, 0.47; 95% CI, 0.40-0.55; P < 0.001). The most common adverse reactions, in order of decreasing frequency, observed in the patients who received TAS-102 were anemia, neutropenia, asthenia/fatigue, nausea, thrombocytopenia, decreased appetite, diarrhea, vomiting, abdominal pain, and pyrexia. Adverse events led to discontinuation of TAS-102 in 3.6% of patients, and 13.7% required a dose reduction. The most common adverse reactions leading to dose reduction were neutropenia, anemia, febrile neutropenia, fatigue, and diarrhea. Clin Cancer Res; 23(12); 2924-7. ©2017 AACR.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Aprobación de Drogas , Resistencia a Antineoplásicos/efectos de los fármacos , Trifluridina/administración & dosificación , Uracilo/análogos & derivados , Adulto , Anciano , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Combinación de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/clasificación , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pirrolidinas , Timina , Trifluridina/efectos adversos , Estados Unidos , United States Food and Drug Administration , Uracilo/administración & dosificación , Uracilo/efectos adversos
4.
Clin Cancer Res ; 22(21): 5171-5176, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27413075

RESUMEN

On December 11, 2015, the FDA granted accelerated approval to alectinib (Alecensa; Genentech) for the treatment of patients with anaplastic lymphoma receptor tyrosine kinase (ALK)-positive, metastatic non-small cell lung cancer (NSCLC) who have progressed on or are intolerant to crizotinib. This approval was based on two single-arm trials including 225 patients treated with alectinib 600 mg orally twice daily. The objective response rates (ORR) by an independent review committee in these studies were 38% [95% confidence interval (CI), 28-49] and 44% (95% CI, 36-53); the median durations of response (DOR) were 7.5 months and 11.2 months. In a pooled analysis of 51 patients with measurable disease in the central nervous system (CNS) at baseline, the CNS ORR was 61% (95% CI, 46-74); the CNS DOR was 9.1 months. The primary safety analysis population included 253 patients. The most common adverse reactions were fatigue (41%), constipation (34%), edema (30%), and myalgia (29%). The most common laboratory abnormalities were anemia (56%), increased aspartate aminotransferase (51%), increased alkaline phosphatase (47%), increased creatine phosphokinase (43%), hyperbilirubinemia (39%), hyperglycemia (36%), increased alanine aminotransferase (34%), and hypocalcemia (32%). Dose reductions due to adverse reactions occurred in 12% of patients, whereas 27% of patients had alectinib dosing interrupted for adverse reactions. Permanent discontinuation of alectinib due to adverse reactions occurred in only 6% of patients. With the clinically meaningful ORR and DOR as well as the safety profile observed in these trials, alectinib was determined to have a favorable benefit-risk profile for the treatment of the indicated population. Clin Cancer Res; 22(21); 5171-6. ©2016 AACR.


Asunto(s)
Carbazoles/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Quinasa de Linfoma Anaplásico , Aspartato Aminotransferasas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Crizotinib , Aprobación de Drogas/métodos , Humanos , Neoplasias Pulmonares/metabolismo , Estados Unidos , United States Food and Drug Administration
5.
Clin Cancer Res ; 22(18): 4545-9, 2016 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-27401247

RESUMEN

On December 11, 2015, the FDA approved uridine triacetate (VISTOGARD; Wellstat Therapeutics Corporation) for the emergency treatment of adult and pediatric patients following a fluorouracil or capecitabine overdose regardless of the presence of symptoms, and of those who exhibit early-onset, severe, or life-threatening toxicity affecting the cardiac or central nervous system, and/or early onset, unusually severe adverse reactions (e.g., gastrointestinal toxicity and/or neutropenia) within 96 hours following the end of fluorouracil or capecitabine administration. Uridine triacetate is not recommended for the nonemergent treatment of adverse reactions associated with fluorouracil or capecitabine because it may diminish the efficacy of these drugs, and the safety and efficacy of uridine triacetate initiated more than 96 hours following the end of administration of these drugs has not been established. The approval is based on data from two single-arm, open-label, expanded-access trials in 135 patients receiving uridine triacetate (10 g or 6.2 g/m(2) orally every 6 hours for 20 doses) for fluorouracil or capecitabine overdose, or who exhibited severe or life-threatening toxicities within 96 hours following the end of fluorouracil or capecitabine administration. Ninety-six percent of patients met the major efficacy outcome measure, which was survival at 30 days or survival until the resumption of chemotherapy, if prior to 30 days. The most common adverse reactions were vomiting, nausea, and diarrhea. This article summarizes the FDA review of this New Drug Application, the data supporting approval of uridine triacetate, and the unique regulatory situations encountered by this approval. Clin Cancer Res; 22(18); 4545-49. ©2016 AACR.


Asunto(s)
Acetatos/farmacología , Acetatos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Aprobación de Drogas , Neoplasias/terapia , Uridina/análogos & derivados , Acetatos/química , Animales , Antineoplásicos/química , Capecitabina/administración & dosificación , Capecitabina/efectos adversos , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Humanos , Neoplasias/diagnóstico , Uso Excesivo de Medicamentos Recetados , Proyectos de Investigación , Resultado del Tratamiento , Estados Unidos , United States Food and Drug Administration , Uridina/química , Uridina/farmacología , Uridina/uso terapéutico
6.
Bioorg Med Chem Lett ; 19(14): 3736-8, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19497744

RESUMEN

We recently reported a beta(3)-decapeptide, betaWWI-1, that binds a validated gp41 model in vitro and inhibits gp41-mediated fusion in cell culture. Here we report six analogs of betaWWI-1 containing a variety of non-natural side chains in place of the central tryptophan of the WWI-epitope. These analogs were compared on the basis of both gp41 affinity in vitro and fusion inibition in live, HIV-infected cells. One new beta(3)-peptide, betaWXI-a, offers a significantly improved CC(50)/EC(50) ratio in the live cell assay.


Asunto(s)
Inhibidores de Fusión de VIH/química , Péptidos/química , Línea Celular , Proteína gp41 de Envoltorio del VIH/metabolismo , Inhibidores de Fusión de VIH/síntesis química , Inhibidores de Fusión de VIH/farmacología , Infecciones por VIH/tratamiento farmacológico , VIH-1 , Humanos , Péptidos/síntesis química , Péptidos/farmacología , Linfocitos T/inmunología , Linfocitos T/metabolismo
7.
J Am Chem Soc ; 127(38): 13126-7, 2005 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-16173723

RESUMEN

Linear peptides derived from the HIV gp41 C-terminus (C-peptides), such as the 36-residue Fuzeon, are potent HIV fusion inhibitors. These molecules bind to the N-peptide region of gp41 and inhibit an intramolecular protein-protein interaction that powers fusion of the viral and host cell membranes. The N-peptide region contains a surface pocket that is occupied in the post-fusion state by three alpha-helical residues found near the gp41 C-terminus: Trp628, Trp631, and Ile635-the WWI epitope. Here, we describe a set of beta3-decapeptides (betaWWI-1-4) in which the WWI epitope is presented on one face of a short 14-helix stabilized by macrodipole neutralization and side chain-side chain salt bridges. betaWWI-1-4 bind in vitro to IZN17, a validated gp41 model, and inhibit syncytia formation in cell culture. Molecules lacking a complete WWI functional epitope neither bind IZN17 nor inhibit syncytia formation. These results provide evidence that short beta-peptide 14-helices can inhibit an intramolecular protein-protein interaction in vivo. Molecules related to betaWWI-1-4 could represent starting points for the development of highly potent inhibitors or antigens effective against HIV or other viruses, including SARS, Ebola, HRSV, and influenza, that employ common fusion mechanisms.


Asunto(s)
Inhibidores de Fusión de VIH/farmacología , VIH-1/efectos de los fármacos , Oligopéptidos/farmacología , Fusión Celular , Inhibidores de Fusión de VIH/química , Células HeLa , Humanos , Oligopéptidos/síntesis química , Oligopéptidos/química
8.
Bioorg Med Chem ; 13(1): 11-6, 2005 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-15582447

RESUMEN

We became interested several years ago in exploring whether 14-helical beta-peptide foldamers could bind protein surfaces and inhibit protein-protein interactions, and if so, whether their affinities and specificities would compare favorably with those of natural or miniature proteins. This exploration was complicated initially by the absence of a suitable beta-peptide scaffold, one that possessed a well-defined 14-helical structure in water and tolerated the diverse sequence variation required to generate high-affinity protein surface ligands. In this perspective, we describe our approach to the design of adaptable beta-peptide scaffolds with high levels of 14-helix structure in water, track the subsequent development of 14-helical beta-peptide protein-protein interaction inhibitors, and examine the potential of this strategy for targeting other therapeutically important proteins.


Asunto(s)
Péptidos/química , Proteínas/antagonistas & inhibidores , Unión Proteica , Proteínas/química
9.
Chem Biol ; 11(9): 1239-50, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15380184

RESUMEN

ADAR2 is an RNA editing enzyme that deaminates adenosines in certain duplex structures. Here, we describe the role of its RNA binding domain, consisting of two copies of a common dsRNA binding motif (dsRBM), in editing site selectivity. ADAR2's dsRBMs bind selectively on a duplex RNA that mimics the Q/R editing site in the glutamate receptor B-subunit pre-mRNA. This selectivity is different from that of PKR's dsRBM I, indicating that dsRBMs from different proteins possess intrinsic binding selectivity. Using directed hydroxyl radical cleavage data, molecular models were developed that predict important recognition surfaces on the RNA for identified dsRBM binding sites. Blocking these surfaces by benzyl modification of guanosine 2-amino groups impeded RNA-editing, demonstrating a correlation between deamination efficiency by ADAR2 and selective binding by its dsRBMs. In addition, the editing activity of a mutant of ADAR2 lacking dsRBM I on N(2)-benzylguanosine-modified RNA suggests the location of the dsRBM I binding site that leads to editing at the GluR-B Q/R site.


Asunto(s)
Adenosina Desaminasa/metabolismo , Guanosina/análogos & derivados , Edición de ARN/fisiología , ARN Bicatenario/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/fisiología , Desaminación , Ensayo de Cambio de Movilidad Electroforética , Guanosina/fisiología , Humanos , Radical Hidroxilo/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica/fisiología , Proteínas de Unión al ARN , Alineación de Secuencia , Especificidad por Sustrato , eIF-2 Quinasa/fisiología
10.
Biopolymers ; 70(1): 86-102, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12925995

RESUMEN

Molecular recognition of double-stranded RNA (dsRNA) is a key event for numerous biological pathways including the trafficking, editing, and maturation of cellular RNA, the interferon antiviral response, and RNA interference. Over the past several years, our laboratory has studied proteins and small molecules that bind dsRNA with the goal of understanding and controlling the binding selectivity. In this review, we discuss members of the dsRBM class of proteins that bind dsRNA. The dsRBM is an approximately 70 amino acid sequence motif found in a variety of dsRNA-binding proteins. Recent results have led to a new appreciation of the ability of these proteins to bind selectivity to certain sites on dsRNA. This property is discussed in light of the RNA selectivity observed in the function of two proteins that contain dsRBMs, the RNA-dependent protein kinase (PKR) and an adenosine deaminase that acts on dsRNA (ADAR2). In addition, we introduce peptide-acridine conjugates (PACs), small molecules designed to control dsRBM-RNA interactions. These intercalating molecules bear variable peptide appendages at opposite edges of an acridine heterocycle. This design imparts the potential to exploit differences in groove characteristics and/or base-pair dynamics at binding sites to achieve selective binding.


Asunto(s)
ARN Bicatenario/química , Acridinas/química , Adenosina Desaminasa/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Ligandos , Microscopía Fluorescente , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Mutación , Conformación de Ácido Nucleico , Péptidos/química , Unión Proteica , Conformación Proteica , Interferencia de ARN , Homología de Secuencia de Aminoácido , Xenopus laevis , eIF-2 Quinasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA