Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Invest New Drugs ; 39(5): 1348-1356, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33830407

RESUMEN

Background We assessed the safety, tolerability, and pharmacokinetics of mitochondrial complex 1 inhibitor ASP4132. Methods This phase I dose-escalation/dose-expansion study enrolled patients with treatment refractory advanced solid tumors to assess safety, dose-limiting toxicities (DLTs), efficacy and pharmacokinetic or oral ASP4132. Results Overall, 39 patients received ASP4132. Acceptable tolerability of ASP4132 5 mg in the first patient led to enrollment in the 10-mg dose cohort. After two DLTs at the 10-mg dose, additional patients were enrolled in the 5-mg cohort; a 7.5-mg cohort and two intermittent-dosing cohorts (ASP4132 10 mg for 3 days, then 4 days off; ASP4132 15 mg for 1 day, then 6 days off). ASP4132 5 mg was well tolerated; however, multiple DLTs such as fatigue, mental status changes, dizziness, lactic acidosis, enteritis, and posterior reversible encephalopathy syndrome were observed in higher dose cohorts (7.5-mg and intermittent 10-mg and 15-mg dose cohorts). Stable disease (+ 4 % to + 15 %) was observed in 8/39 (20.5 %) patients. ASP4132 plasma pharmacokinetics were characterized by high variability, with rapid absorption and accumulation from slow elimination. Conclusions ASP4132 showed limited clinical activity, and DLTs prohibited dose escalation. Further research is required to determine if DLTs will limit clinical activity of other mitochondrial complex I inhibitors. Clinical Trial ID (clinicaltrials.gov): NCT02383368, March 9, 2015.


Asunto(s)
Antineoplásicos/uso terapéutico , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Piperazinas/uso terapéutico , Piridinas/uso terapéutico , Adulto , Anciano , Antineoplásicos/efectos adversos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Piperazinas/efectos adversos , Piridinas/efectos adversos
2.
Invest New Drugs ; 38(2): 445-456, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31041575

RESUMEN

ASP5878 is a selective small-molecule inhibitor of fibroblast growth factor receptors (FGFRs). This study investigated safety, tolerability, and antitumor effect of single and multiple oral doses of ASP5878 in patients with solid tumors. This phase 1, open label, first-in-human study comprised dose-escalation and dose-expansion parts. Primary objectives of the dose-escalation part were to identify the dose-limiting toxicity (DLT), maximum tolerated dose, and recommended dose of ASP5878 for the dose-expansion part. Nine dose cohorts of ASP5878 were evaluated (0.5─2 mg once daily; 2─40 mg twice daily [BID]). A single dose of ASP5878 was followed by a 2-day pharmacokinetic collection, and then either 28-day cycles of daily dosing (ASP5878 ≤ 10 mg BID) or 5-day dosing/2-day interruption (ASP5878 ≥ 20 mg BID). The primary objective of the dose-expansion part was to determine the safety of ASP5878 (16 mg BID) administered in 28-day cycles of 5-day dosing/2-day interruption in patients with urothelial carcinoma, hepatocellular carcinoma, or squamous cell lung carcinoma with FGFR genetic alterations. Safety was assessed by monitoring adverse events (AEs). Thirty-five patients were enrolled and 31 discontinued in the dose-escalation part; 51 patients were enrolled and 51 discontinued in the dose-expansion part. In the dose-escalation part, 66.7% of patients in the 20 mg BID 5-day dosing/2-day interruption group reported DLTs of hyperphosphatemia. The recommended dose for the dose-expansion part was 16 mg BID. Common AEs included retinal detachment, diarrhea, and increased alanine aminotransferase. One death occurred that was not related to ASP5878. ASP5878 was well tolerated with manageable toxicities including hyperphosphatemia.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias de Células Escamosas/tratamiento farmacológico , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Neoplasias Urológicas/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Carcinoma Hepatocelular/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias de Células Escamosas/metabolismo , Hormona Paratiroidea/sangre , Fosfatos/sangre , Pirazoles/efectos adversos , Pirazoles/sangre , Pirazoles/farmacocinética , Pirimidinas/efectos adversos , Pirimidinas/sangre , Pirimidinas/farmacocinética , Resultado del Tratamiento , Neoplasias Urológicas/metabolismo , Adulto Joven
3.
J Hematol Oncol ; 9: 23, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26966027

RESUMEN

BACKGROUND: ASP3026 is a second-generation anaplastic lymphoma kinase (ALK) inhibitor that has potent in vitro activity against crizotinib-resistant ALK-positive tumors. This open-label, multicenter, first-in-human phase I study ( NCT01284192 ) assessed the safety, pharmacokinetic profile, and antitumor activity of ASP3026. METHODS: Advanced solid tumor patients received oral ASP3026 in 3 + 3 dose-escalation cohorts at doses of 25-800 mg once daily in 28-day cycles. The endpoints were to identify the maximum tolerated dose (MTD), the recommended phase II dose (RP2D), and the pharmacokinetic profile of ASP3026. A phase Ib expansion cohort enrolled patients with metastatic, crizotinib-resistant ALK-positive solid tumors at the RP2D, and response was evaluated by RECIST 1.1. RESULTS: The dose-escalation cohort enrolled 33 patients, including three crizotinib-resistant, ALK-positive patients, and the dose-expansion cohort enrolled another 13 crizotinib-resistant, ALK-positive non-small cell lung cancer (NSCLC) patients. ASP3026 demonstrated both linear pharmacokinetics and dose-proportional exposure for area under the plasma concentration-time curve and maximum concentration observed with a median terminal half-life of 35 h, supporting the daily dosing. Grade 3 rash and elevated transaminase concentrations were dose-limiting toxicities observed at 800 mg; hence, 525 mg daily was the MTD and RP2D. The most common treatment-related adverse events were nausea (38%), fatigue (35%), and vomiting (35 %). Among the 16 patients with crizotinib-resistant ALK-positive tumors (15 NSCLC, 1 neuroblastoma), eight patients achieved partial response (overall response rate 50%; 95% confidence interval 25-75%) and seven patients (44%) achieved stable disease. CONCLUSIONS: ASP3026 was well tolerated and had therapeutic activity in patients with crizotinib-resistant ALK-positive advanced tumors. TRIAL REGISTRATION: ClinTrials.gov: NCT01284192.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Sulfonas/administración & dosificación , Triazinas/administración & dosificación , Administración Oral , Adulto , Anciano , Quinasa de Linfoma Anaplásico , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Área Bajo la Curva , Crizotinib , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Resistencia a Antineoplásicos/efectos de los fármacos , Fatiga/inducido químicamente , Femenino , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Náusea/inducido químicamente , Neoplasias/enzimología , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Sulfonas/efectos adversos , Sulfonas/farmacocinética , Resultado del Tratamiento , Triazinas/efectos adversos , Triazinas/farmacocinética , Vómitos/inducido químicamente , Adulto Joven
4.
Clin Cancer Res ; 22(20): 5058-5067, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27401244

RESUMEN

PURPOSE: Tivozanib, a selective inhibitor of VEGFR-1, -2, and -3, plus mFOLFOX6 in an advanced gastrointestinal cancer phase Ib study had encouraging antineoplastic activity and a tolerable safety profile. This randomized, open-label, phase II trial of tivozanib/mFOLFOX6 versus bevacizumab/mFOLFOX6 in patients with previously untreated metastatic colorectal cancer (mCRC) evaluated tivozanib activity versus bevacizumab. EXPERIMENTAL DESIGN: Treatment-naïve patients received mFOLFOX6 every 2 weeks of each 28-day cycle plus either tivozanib orally 1.5 mg once daily for 21 days or bevacizumab intravenously 5 mg/kg every 2 weeks. Investigator-assessed progression-free survival (PFS) was the primary endpoint; some secondary endpoints included safety, overall survival, overall response rate (ORR), duration of response, time to treatment failure, and biomarker subgroup analyses. RESULTS: A prespecified interim futility analysis demonstrated that the futility boundary for superiority of tivozanib/mFOLFOX6 over bevacizumab/mFOLFOX6 for PFS in the intent-to-treat population was crossed; median PFS was 9.4 versus 10.7 months [HR = 1.091; confidence interval (CI), 0.693-1.718; P = 0.706]. Tivozanib/mFOLFOX6 resulted in PFS and ORR comparable with bevacizumab/mFOLFOX6; interim analyses biomarker results revealed no significant PFS association. Post hoc final analyses demonstrated a potential difference in tivozanib-specific PFS in patients with low neuropilin-1 (NRP-1), but not in patients with high NRP-1. Tivozanib/mFOLFOX6 was tolerable and adverse events were comparable with both bevacizumab/mFOLFOX6 and previous tivozanib studies. CONCLUSIONS: The efficacy of tivozanib/mFOLFOX6 was comparable with but not superior to bevacizumab/mFOLFOX6 in patients with previously untreated mCRC. Since data from the prespecified interim analysis did not demonstrate superiority, this resulted in discontinuation of the study. The safety and tolerability profile of tivozanib/mFOLFOX6 was consistent with other tivozanib trials. NRP-1 is a potential predictive biomarker for tivozanib activity, but these results require further validation. Clin Cancer Res; 22(20); 5058-67. ©2016 AACR.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bevacizumab/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Compuestos de Fenilurea/uso terapéutico , Quinolinas/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bevacizumab/efectos adversos , Biomarcadores de Tumor/sangre , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/efectos adversos , Fluorouracilo/uso terapéutico , Humanos , Leucovorina/efectos adversos , Leucovorina/uso terapéutico , Masculino , Persona de Mediana Edad , Neuropilina-1/sangre , Compuestos Organoplatinos/efectos adversos , Compuestos Organoplatinos/uso terapéutico , Compuestos de Fenilurea/efectos adversos , Quinolinas/efectos adversos
5.
J Clin Oncol ; 21(21): 3955-64, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14517187

RESUMEN

PURPOSE: PTK787/ZK 222584 (PTK/ZK), an orally active inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinases, inhibits VEGF-mediated angiogenesis. The pharmacodynamic effects of PTK/ZK were evaluated by assessing changes in contrast-enhancement parameters of metastatic liver lesions using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in patients with advanced colorectal cancer treated in two ongoing, dose-escalating phase I studies. PATIENTS AND METHODS: Twenty-six patients had DCE-MRI performed at baseline, day 2, and at the end of each 28-day cycle. Doses of oral PTK/ZK ranged from 50 to 2000 mg once daily. Tumor permeability and vascularity were assessed by calculating the bidirectional transfer constant (Ki). The percentage of baseline Ki (% of baseline Ki) at each time point was compared with pharmacokinetic and clinical end points. RESULTS: A significant negative correlation exists between the % of baseline Ki and increase in PTK/ZK oral dose and plasma levels (P =.01 for oral dose; P =.0001 for area under the plasma concentration curve at day 2). Patients with a best response of stable disease had a significantly greater reduction in Ki at both day 2 and at the end of cycle 1 compared with progressors (mean difference in % of baseline Ki, 47%, P =.004%; and 51%, P =.006; respectively). The difference in % of baseline Ki remained statistically significant after adjusting for baseline WHO performance status. CONCLUSION: These findings should help to define a biologically active dose of PTK/ZK. These results suggest that DCE-MRI may be a useful biomarker for defining the pharmacological response and dose of angiogenesis inhibitors, such as PTK/ZK, for further clinical development.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Neoplasias Colorrectales/sangre , Neoplasias Hepáticas/sangre , Imagen por Resonancia Magnética/normas , Ftalazinas/farmacocinética , Piridinas , Administración Oral , Adulto , Anciano , Inhibidores de la Angiogénesis/administración & dosificación , Área Bajo la Curva , Biomarcadores , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Medios de Contraste , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Compuestos Organometálicos , Ftalazinas/administración & dosificación , Valor Predictivo de las Pruebas , Resultado del Tratamiento
6.
J Med Chem ; 46(16): 3508-13, 2003 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-12877589

RESUMEN

In a program aimed at the development of neurokinin antagonists, N-[(R,R)-(E)-1-(3,4-dichlorobenzyl)-3-(2-oxoazepan-3-yl)carbamoyl]allyl-N-methyl-3,5-bis(trifluoromethyl)benzamide (1, DNK333) has been discovered as a potent and balanced neurokinin (tachykinin) NK(1)/NK(2) receptor antagonist. Enantiomerically pure (>99.5% ee) 1 can be prepared in 6 + 1 synthetic steps starting from commercially available optically active BOC-d-3,4-dichlorophenylalanine in an overall yield of ca. 25-30%. 1 showed potent affinities to cloned human NK(1) (pK(i) = 8.38) and NK(2) (pK(i) = 8.02) receptors. When 1 was compared to the other possible three diastereoisomers, it could be demonstrated that only the R,R-isomer (1) exhibits potent and balanced affinity for the cloned human NK(1) and NK(2) receptors. 1 exhibited favorable pharmacokinetic properties in guinea pigs following oral administration and demonstrated in vivo activity in pharmacological models of substance P- and neurokinin A (NKA)-induced bronchoconstriction in guinea pigs after intravenous and in squirrel monkeys after oral application.


Asunto(s)
Compuestos Aza/síntesis química , Benzamidas/síntesis química , Antagonistas del Receptor de Neuroquinina-1 , Receptores de Neuroquinina-2/antagonistas & inhibidores , Administración Oral , Animales , Compuestos Aza/química , Compuestos Aza/farmacología , Benzamidas/química , Benzamidas/farmacología , Disponibilidad Biológica , Broncoconstricción/efectos de los fármacos , Células CHO , Cricetinae , Cobayas , Humanos , Masculino , Saimiri , Estereoisomerismo , Relación Estructura-Actividad
7.
Am J Pharmacogenomics ; 3(2): 97-106, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12749727

RESUMEN

The human genome project, as well as advances in our understanding of asthma susceptibility, are yielding novel candidate targets for disease intervention. The normalization of up-regulated gene expression may treat or improve the disease outcome. However, only some of these gene product targets may be 'tractable', i.e. amenable to blockade by small, orally active, organic molecules. The remainder have been termed 'non-tractable'. For over a decade, antisense oligonucleotides (ASONs) have been used as tools to evaluate the importance of specific gene products in vitro. In recent years evidence has accumulated indicating their potential as a viable new therapeutic approach in their own right, being able to block 'non-tractable' targets as well as 'tractable' targets.Distribution, cell-specific uptake, and effectiveness of aerosolized phosphorothioate ASONs are currently being evaluated in animal models. The results demonstrate broad distribution throughout the lung, and uptake by all of the cell types examined to date. Functionality has been demonstrated against diverse targets, including nuclear transcription factors, tyrosine kinases, G-protein coupled receptors, cytokine receptors, growth factors, and chemokines.EPI-2010, a respirable ASON (RASON) against the adenosine A(1) receptor, is the first test case for this new class of respiratory therapeutics. The rationale for EPI-2010 is that overactivity of the adenosine-signaling pathway in asthmatic lungs contributes to airway inflammation and hyperresponsiveness. EPI-2010 binds to the initiation codon of the adenosine A(1) receptor mRNA, and thereby blocks translation and targets the message for degradation by RNase. EPI-2010 is apparently metabolized locally by endogenous nucleases confining its activity to the airways. Phase I clinical trials have shown EPI-2010 to be well-tolerated, with indications of efficacy. In conclusion, one important application of RASONs is in addressing up-regulated disease targets, only some of which are 'tractable' by small molecules. It is hoped that this will yield new therapeutic options to the benefit of patients with asthma and allergic disorders.


Asunto(s)
Asma/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Oligonucleótidos Antisentido/administración & dosificación , Administración por Inhalación , Animales , Asma/inmunología , Humanos
8.
Clin Rev Allergy Immunol ; 27(3): 207-17, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15630157

RESUMEN

Advances in our understanding of asthma pathogenesis and delineation of the human genome project are yielding novel candidate targets for therapeutic intervention. In parallel with target identification, the past decade has produced novel approaches to normalizing expression genes that are upregulated in disease processes. Single-stranded antisense oligonucleotides and double-stranded short-interfering RNA molecules, which specifically mark target transcripts for degradation, are being investigated for their ability to modulate disease processes. In both cases, the targets are RNA transcripts, and not protein; therefore, all types of molecular gene products can be inhibited, including historically undrugable species such as transcription factors and phosphatases. Various RNA interference strategies have been successfully tested in vitro and in animal models of disease, and data is beginning to accumulate from human clinical trials.EPI-2010, a 21-mer phosphorothioate against the adenosine A1 receptor promoter region,has completed preclinical pharmacology testing and its initial clinical trials. The rationale for EPI-2010 is that overactivity of the adenosine signaling pathway in asthmatic lungs contributes to airway inflammation and hyperresponsiveness. Phase I/IIa clinical trials have shown EPI-2010 to be safe and well-tolerated, with modest indications of efficacy in patients with mild asthma.


Asunto(s)
Asma/terapia , Hipersensibilidad/terapia , Oligonucleótidos Antisentido/uso terapéutico , Interferencia de ARN , Asma/tratamiento farmacológico , Humanos , Hipersensibilidad/tratamiento farmacológico
9.
Cancer Chemother Pharmacol ; 71(1): 93-101, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23064954

RESUMEN

PURPOSE: Pazopanib plus gemcitabine combination therapy was explored in patients with advanced solid tumors. METHODS: In a modified 3 + 3 enrollment scheme, oral once-daily pazopanib was administered with intravenous gemcitabine (Days 1 and 8, 21-day cycles). Three protocol-specified dose levels were tested: pazopanib 400 mg plus gemcitabine 1,000 mg/m(2), pazopanib 800 mg plus gemcitabine 1,000 mg/m(2), and pazopanib 800 mg plus gemcitabine 1,250 mg/m(2). Maximum-tolerated dose was based on dose-limiting toxicities during treatment Cycle 1. In the expansion phase, six additional patients were enrolled at the highest tolerable dose level. RESULTS: Twenty-two patients were enrolled. At the highest dose level tested (pazopanib 800 plus gemcitabine 1,250), patients received >80% of their planned dose and the regimen was deemed safe and tolerable. The most common treatment-related adverse events included fatigue, neutropenia, nausea, and decreased appetite. Neutropenia and thrombocytopenia were the most common events leading to dose modifications. Pharmacokinetic interaction between pazopanib and gemcitabine was not observed. One objective partial response at the highest dose was observed in a patient with metastatic melanoma. Prolonged disease stabilization (>12 cycles) was reported in three patients (metastatic melanoma, cholangiocarcinoma, and colorectal carcinoma). CONCLUSION: Combination pazopanib plus gemcitabine therapy is tolerable, with an adverse event profile reflective of that associated with the individual agents. There was no apparent pharmacokinetic interaction with pazopanib plus gemcitabine co-administration, although patient numbers were limited. Further investigation of combined pazopanib plus gemcitabine is warranted.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Humanos , Indazoles , Infusiones Intravenosas , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/patología , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Resultado del Tratamiento , Adulto Joven , Gemcitabina
10.
Cancer Chemother Pharmacol ; 71(3): 565-73, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23344712

RESUMEN

PURPOSE: As tyrosine kinase inhibitors have been associated with cardiotoxicity, we evaluated the effect of pazopanib, an inhibitor of vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and c-Kit, on electrocardiographic parameters in patients with cancer. METHODS: This double-blind, placebo-controlled, parallel-group study randomized patients (N = 96) to moxifloxacin (positive control) or placebo on Day 1 followed by pazopanib or placebo 800 mg/day (fasted) on Days 2-8 and 1,600 mg (with food) on Day 9. Treatment effects were evaluated by baseline-adjusted, time-matched, serial Holter electrocardiograms. RESULTS: Sixty-five patients were evaluable for preplanned analyses. On Day 1, the maximum mean difference in baseline-adjusted, time-matched Fridericia-corrected QT (QTcF) interval in moxifloxacin-treated patients versus placebo was 10.6 ms (90% confidence interval [CI]: 4.2, 17.0). The administration scheme increased plasma pazopanib concentrations approximately 1.3- to 1.4-fold versus the recommended 800 mg once-daily dose. Pazopanib caused clinically significant increases from baseline in blood pressure, an anticipated class effect, and an unexpected reduction in heart rate from baseline that correlated with pazopanib exposure. On Day 9, the maximum mean difference in baseline-adjusted, time-matched QTcF interval in pazopanib-treated patients versus placebo was 4.4 ms (90% CI: -2.4, 11.2). Mixed-effects modeling indicated no significant concentration-dependent effect of pazopanib or its metabolites on QTcF interval. CONCLUSIONS: Pazopanib as administered in this study achieved supratherapeutic concentrations, produced a concentration-dependent decrease in heart rate, and caused a small, concentration-independent prolongation of the QTcF interval.


Asunto(s)
Inhibidores de la Angiogénesis/efectos adversos , Sistema de Conducción Cardíaco/efectos de los fármacos , Neoplasias/complicaciones , Pirimidinas/efectos adversos , Sulfonamidas/efectos adversos , Adulto , Anciano , Algoritmos , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacocinética , Antibacterianos/farmacocinética , Compuestos Aza/farmacocinética , Presión Sanguínea/efectos de los fármacos , Intervalos de Confianza , Método Doble Ciego , Electrocardiografía Ambulatoria , Femenino , Fluoroquinolonas , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Indazoles , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/fisiopatología , Masculino , Persona de Mediana Edad , Moxifloxacino , Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Quinolinas/farmacocinética , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacocinética , Adulto Joven
11.
Mol Cancer Ther ; 11(8): 1820-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22679111

RESUMEN

Several phase III trials have shown that the addition of an antiangiogenic agent to conventional chemotherapy can improve clinical benefit in patients with advanced solid tumors. This study examined the feasibility of combining pazopanib (Votrient), an oral antiangiogenic agent, with paclitaxel and carboplatin. This 3 + 3 dose-escalation phase I study evaluated the maximum-tolerated regimen (MTR) of daily pazopanib in combination with paclitaxel 175 mg/m(2) and carboplatin [dosed at area under the curve (AUC) 5 or 6] given every 21 days in patients with advanced solid tumors. Plasma samples were collected to evaluate the effect of pazopanib on the pharmacokinetics of paclitaxel and carboplatin. Thirty-four patients were enrolled. The MTR was paclitaxel 175 mg/m(2) and carboplatin AUC5 with pazopanib 200 mg. The most common dose-limiting toxicities were neutropenia and thrombocytopenia. Two patients with esophageal cancer had a complete response and four patients, one each with breast, small-cell lung, pancreatic, and gastroesophageal junction cancer, had partial responses. Pazopanib at 200 mg increased paclitaxel maximal concentration (C(max)) by 43% and carboplatin (AUC5 or AUC6) C(max) by 54%. Paclitaxel and carboplatin given every 21 days at standard doses was not feasible in combination with the monotherapy pazopanib dose of 800 mg daily because of dose-limiting myelosuppression. Coadministration of pazopanib increased exposure to paclitaxel and carboplatin and likely contributed to this effect. Given the antitumor activity of this regimen, further studies are underway to determine a clinically tolerable schedule of pazopanib with paclitaxel and carboplatin.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carboplatino/administración & dosificación , Femenino , Humanos , Indazoles , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Paclitaxel/administración & dosificación , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Resultado del Tratamiento
12.
J Clin Oncol ; 29(18): 2557-64, 2011 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-21576632

RESUMEN

PURPOSE: Pazopanib, an oral angiogenesis inhibitor, is approved for the treatment of advanced renal cell carcinoma (RCC). Response to pazopanib monotherapy varies between patients, and no validated biomarkers predictive of treatment outcome have been identified. We tested the hypothesis that this variability is partially dependent on germline genetic variants that may affect pazopanib exposure or angiogenesis pathways. PATIENTS AND METHODS: Twenty-seven functional polymorphisms within 13 genes were evaluated in 397 patients with RCC. Genetic association with progression-free survival (PFS) and objective response rate (RR) was analyzed using the Cox proportional hazards model and proportional odds model, respectively. RESULTS: Three polymorphisms in IL8 and HIF1A and five polymorphisms in HIF1A, NR1I2, and VEGFA showed nominally significant association (P ≤ .05) with PFS and RR, respectively. Compared with the wild-type AA genotype (median PFS, 48 weeks), the IL8 2767TT variant genotype showed inferior PFS (27 weeks, P = .009). The HIF1A 1790AG genotype was associated with inferior PFS and reduced RR, compared with the wild-type GG genotype (median PFS, 20 v 44 weeks; P = .03; RR, 30% v 43%, P = .02). Reductions in RR were detected for the NR1I2 -25385TT genotype, compared with the wild-type CC genotype (37% v 50%, P = .03), and for the VEGFA -1498CC genotype compared with the TT genotypes (33% v 51%). CONCLUSION: Germline variants in angiogenesis- and exposure-related genes may predict treatment response to pazopanib monotherapy in patients with RCC. If validated, these markers may explain why certain patients fail antiangiogenesis therapy and they may support the use of alternative strategies to circumvent this issue.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Proteínas de Neoplasias/genética , Neovascularización Fisiológica/genética , Polimorfismo de Nucleótido Simple , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Inhibidores de la Angiogénesis/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Ensayos Clínicos Fase II como Asunto/estadística & datos numéricos , Estudios Cruzados , Análisis Mutacional de ADN , Supervivencia sin Enfermedad , Marcadores Genéticos , Genotipo , Mutación de Línea Germinal , Humanos , Indazoles , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Estudios Multicéntricos como Asunto/estadística & datos numéricos , Modelos de Riesgos Proporcionales , Pirimidinas/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Sulfonamidas/farmacología
13.
Antisense Nucleic Acid Drug Dev ; 12(3): 177-81, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12162700

RESUMEN

Respirable antisense oligonucleotides (RASONs) represent a novel class of respiratory therapeutic molecules with the potential to specifically address the challenges posed by the successes of the Human Genome Program, namely, the need to rapidly identify the critical pulmonary disease-relevant drugable targets from the vast pool of 30,000-40,000 human genes and to discover and develop drugs that specifically attack these targets. We have shown that EPI-2010, a RASON targeting the adenosine A1 receptor, a G-protein coupled receptor that has been implicated in the regulation of three major determinants of asthma, can be delivered directly to the target disease tissue as an aerosol formulation. In vivo efficacy, absorption, distribution, metabolism, and excretion (ADME), and safety studies of inhaled EPI-2010 employing animal models of human asthma suggest that the RASON approach enables the specific delivery of efficacious, safe, and long-acting doses of phosphorothioate oligonucleotides to the respiratory tract. Moreover, these data indicate that RASONs truly have the potential to address the respiratory drug discovery bottleneck of the postgenomic era, that is, the ability to rapidly validate disease targets and develop pulmonary disease therapeutics for these validated targets.


Asunto(s)
Asma/tratamiento farmacológico , Diseño de Fármacos , Oligonucleótidos Antisentido , Fosfatos , Administración por Inhalación , Sistema Cardiovascular/efectos de los fármacos , Humanos , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/efectos adversos , Oligonucleótidos Antisentido/síntesis química , Oligonucleótidos Antisentido/farmacocinética , Oligonucleótidos Antisentido/uso terapéutico , Fosfatos/administración & dosificación , Fosfatos/efectos adversos , Fosfatos/farmacocinética , Fosfatos/uso terapéutico , Distribución Tisular
14.
Bioorg Med Chem Lett ; 12(16): 2065-8, 2002 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12127505

RESUMEN

In a continuation of our efforts to simplify the structure of our neurokinin antagonists, a series of substituted biphenyl derivatives has been prepared. Several compounds exhibit potent affinities for both the NK(1) receptor (<10nM) and for the NK(2) receptor (<50 nM). Details on the design, synthesis, biological activities, SAR and conformational analysis of this new class of dual NK(1)/NK(2) receptor antagonists are presented.


Asunto(s)
Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Antagonistas del Receptor de Neuroquinina-1 , Receptores de Neuroquinina-2/antagonistas & inhibidores , Animales , Broncoconstricción/efectos de los fármacos , Células CHO , Cricetinae , Cobayas , Humanos , Conformación Molecular , Estructura Molecular , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA