Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Invest New Drugs ; 35(5): 576-588, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28424891

RESUMO

Background Combinations of molecularly targeted agents may provide optimal anti-tumor activity and improve clinical outcomes for patients with advanced cancers. Selumetinib (AZD6244, ARRY-142886) is an oral, potent and highly selective, allosteric inhibitor of MEK1/2, a component of the RAS/RAF/MEK/ERK pathway which is constitutively activated in many cancers. We investigated the safety, tolerability, and pharmacokinetics (PK) of selumetinib in combination with molecularly targeted drugs erlotinib or temsirolimus in patients with advanced solid tumors. Methods Two-part study: dose escalation, to determine the maximum tolerated dose (MTD) of selumetinib in combination with erlotinib 100 mg once daily (QD) or temsirolimus 25 mg once weekly, followed by dose expansion at the respective combination MTDs to further investigate safety and anti-tumor effects. Results 48 patients received selumetinib plus erlotinib and 32 patients received selumetinib plus temsirolimus. The MTD with erlotinib 100 mg QD was selumetinib 100 mg QD, with diarrhea being dose limiting. The most common all grade adverse events (AEs): diarrhea, rash, nausea, and fatigue. Four (8.3%) patients had ≥12 weeks stable disease. The MTD with temsirolimus 25 mg once weekly was selumetinib 50 mg twice daily (BID), with mucositis and neutropenia being dose limiting. The most commonly reported AEs: nausea, fatigue, diarrhea, and mucositis. Ten (31.3%) patients had ≥12 weeks stable disease. The combination PK profiles were comparable to previously observed monotherapy profiles. Conclusions MTDs were established for selumetinib in combination with erlotinib or temsirolimus. Overlapping toxicities prevented the escalation of selumetinib to its recommended phase II monotherapy dose of 75 mg BID. TRIAL REGISTRATION: ClinicalTrials.gov NCT00600496; registered 8 July 2009.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Antineoplásicos/administração & dosagem , Benzimidazóis/administração & dosagem , Relação Dose-Resposta a Droga , Cloridrato de Erlotinib/administração & dosagem , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/administração & dosagem , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados
3.
BMC Cancer ; 17(1): 173, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28264648

RESUMO

BACKGROUND: The RAS/RAF/MEK/ERK pathway is constitutively activated in many cancers. Selumetinib (AZD6244, ARRY-142886) is an oral, potent and highly selective, allosteric MEK1/2 inhibitor with a short half-life that has shown clinical activity as monotherapy in phase I and II studies of advanced cancer. Preclinical data suggest that selumetinib may enhance the activity of chemotherapeutic agents. We assessed the safety, tolerability, and pharmacokinetics (PK) of selumetinib (AZD6244, ARRY-142886) in combination with docetaxel or dacarbazine in patients with advanced solid tumors. METHODS: This study was a phase I, open-label, multicenter study in patients aged ≥18 years with advanced solid tumors who were candidates for docetaxel or dacarbazine treatment. Part A of the study (dose escalation) evaluated safety, tolerability, PK, and maximum tolerated dose (MTD) of selumetinib twice daily (BID) with docetaxel 75 mg/m2 or dacarbazine 1000 mg/m2 administered every 21 days. Patients receiving docetaxel could be administered primary prophylactic granulocyte-colony stimulating factor according to standard guidelines. Part B of the study (dose expansion) further evaluated safety, tolerability, and PK in 12 additional patients at the MTD combinations determined in part A. RESULTS: A total of 35 patients received selumetinib plus docetaxel, and 25 received selumetinib plus dacarbazine. The MTD of selumetinib was 75 mg BID in combination with either docetaxel (two dose-limiting toxicity [DLT] events: neutropenia with fever, and thrombocytopenia) or dacarbazine (one DLT event: thrombocytopenia). Common adverse events occurring with each treatment combination were diarrhea, peripheral/periorbital edema, fatigue, and nausea. PK parameters for selumetinib and docetaxel or dacarbazine were similar when administered alone or in combination. Partial responses were reported in 6/35 patients receiving selumetinib plus docetaxel and 4/25 patients receiving selumetinib plus dacarbazine. CONCLUSIONS: The combinations of selumetinib plus docetaxel and selumetinib plus dacarbazine demonstrated manageable safety and tolerability profiles and preliminary signs of clinical activity in patients with advanced solid tumors. TRIAL REGISTRATION: ClinicalTrials.gov NCT00600496; registered 8 July 2009.


Assuntos
Benzimidazóis/efeitos adversos , Dacarbazina/efeitos adversos , Neoplasias/tratamento farmacológico , Taxoides/efeitos adversos , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzimidazóis/farmacocinética , Benzimidazóis/uso terapêutico , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Docetaxel , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Segurança do Paciente , Taxoides/farmacocinética , Taxoides/uso terapêutico
4.
Oncologist ; 18(7): 819-20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23847256

RESUMO

BACKGROUND: AZD1480 is a novel agent that inhibits Janus-associated kinases 1 and 2 (JAK1 and JAK2). The primary objective of this phase I study was to investigate the safety and tolerability of AZD1480 when administered as monotherapy to patients with solid tumors. METHODS: Thirty-eight patients with advanced malignancies were treated at doses of 10-70 mg once daily (QD) and 20-45 mg b.i.d. RESULTS: Pharmacokinetic (PK) analysis revealed rapid absorption and elimination with minimal accumulation after repeated QD or b.i.d. dosing. Exposure increased in a dose-dependent manner from 10-50 mg. Maximum plasma concentration (Cmax) was attained ∼1 hour after dose, and t1/2 was ∼5 hours. Pharmacodynamic analysis of circulating granulocytes demonstrated maximum phosphorylated STAT3 (pSTAT3) inhibition 1-2 hours after dose, coincident with Cmax, and greater pSTAT3 inhibition at higher doses. The average pSTAT3 inhibition in granulocytes at the highest dose tested, 70 mg QD, was 56% (standard deviation: ±21%) at steady-state drug levels. Dose-limiting toxicities (DLTs) consisted of pleiotropic neurologic adverse events (AEs), including dizziness, anxiety, ataxia, memory loss, hallucinations, and behavior changes. These AEs were generally reversible with dose reduction or treatment cessation. CONCLUSIONS: Whether the DLTs were due to inhibition of JAK-1/2 or to off-target effects is unknown. The unusual DLTs and the lack of clinical activity led to discontinuation of development.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Relação Dose-Resposta a Droga , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/classificação , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Humanos , Janus Quinase 2/genética , Masculino , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais
5.
Cancer Prev Res (Phila) ; 4(3): 311-23, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21372031

RESUMO

This article endeavors to clarify the current requirements and status of regulatory approval for chemoprevention (risk reduction) drugs and discusses possible improvements to the regulatory pathway for chemoprevention. Covering a wide range of topics in as much depth as space allows, this report is written in a style to facilitate the understanding of nonscientists and to serve as a framework for informing the directions of experts engaged more deeply with this issue. Key topics we cover here are as follows: a history of definitive cancer chemoprevention trials and their influence on the evolution of regulatory assessments; a brief review of the long-standing success of pharmacologic risk reduction of cardiovascular diseases and its relevance to approval for cancer risk reduction drugs; the use and limitations of biomarkers for developing and the approval of cancer risk reduction drugs; the identification of individuals at a high(er) risk for cancer and who are appropriate candidates for risk reduction drugs; business models that should incentivize pharmaceutical industry investment in cancer risk reduction; a summary of scientific and institutional barriers to development of cancer risk reduction drugs; and a summary of major recommendations that should help facilitate the pathway to regulatory approval for pharmacologic cancer risk reduction drugs.


Assuntos
Anticarcinógenos/farmacologia , Quimioprevenção/métodos , Aprovação de Drogas , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Ensaios Clínicos como Assunto , Indústria Farmacêutica/métodos , Detecção Precoce de Câncer , Feminino , Humanos , Masculino , Oncologia/métodos , Neoplasias/prevenção & controle , Cooperação do Paciente , Risco
10.
Oncologist ; 8 Suppl 1: 27-30, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12626786

RESUMO

Research in cancer-related fatigue lags far behind research in cancer-related pain and is astonishingly underdeveloped given the magnitude of the problem among cancer patients. This was recently recognized at the State-of-the-Science Conference on Symptom Management in Cancer: Pain, Depression, and Fatigue, held at the U.S. National Institutes of Health in July 2002. The results of patient surveys on fatigue being conducted in Ireland by the All Ireland Fatigue Coalition (AIFC) closely mirror results from similar surveys conducted in the U.S., in some regards, with 53% of patients experiencing significant fatigue at least daily, and 80% at least monthly on both sides of the Atlantic. In other respects, such as duration of bouts of fatigue, how much and with whom patients discuss their fatigue symptoms, and economic impacts of fatigue, there are significant differences between survey findings in the U.S. and in Ireland. But in both places, the condition is underreported and undertreated and takes an enormous toll. Work by the AIFC to document the problem in Ireland, expand public awareness, and conduct research on which to base guidelines for diagnosis and treatment is an excellent beginning to tackling this long understudied condition that afflicts the overwhelming majority of cancer patients.


Assuntos
Fadiga/etiologia , Neoplasias/complicações , Inquéritos Epidemiológicos , Humanos , Irlanda , Neoplasias/terapia , Qualidade de Vida , Estados Unidos
11.
Oncologist ; 7(5): 401-9, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12401902

RESUMO

Anticancer drug discovery has shifted from an empiric random screening directed approach to a more rational and mechanistic, target-based approach, which reflects our rapidly expanding knowledge of the pathogenesis of a variety of forms of cancer at the molecular level, providing new targets for drug discovery and development. The clinical development of target-based anticancer drugs will require fundamental changes to the traditional clinical trial design and end points that have been used for conventional cytotoxic drugs. In the phase I and II settings, traditional end points (toxicity and response) may not be suitable for more selective, cytostatic target-based agents, and these end points may be replaced by biological or pharmacokinetic end points to define the optimal doses and the therapeutic effects of these drugs on their targets. For phase III trials, measurable clinical benefit will continue to be the primary end point. As our understanding of the complex pathways and networks controlling cell signaling, proliferation, and cell death expands, we must learn how and when to use agents to target specific steps in malignant transformation and proliferation, and we must adapt clinical trial design to test the clinical utility of this promising new class of anticancer drugs.


Assuntos
Antineoplásicos/farmacologia , Ensaios Clínicos como Assunto/normas , Desenho de Fármacos , Neoplasias/tratamento farmacológico , Projetos de Pesquisa , Avaliação de Medicamentos/métodos , Tratamento Farmacológico/tendências , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...