Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Acta Oncol ; 61(1): 52-57, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34736367

RESUMEN

INTRODUCTION: METEOR was a phase 3 trial (NCT01865747) of cabozantinib versus everolimus in adults with advanced or metastatic clear cell RCC previously treated with VEGF receptor (VEGFR) tyrosine kinase inhibitors (TKIs). This post hoc analysis of METEOR compared outcomes for patients recruited from European and non-European countries. MATERIAL AND METHODS: Adults with advanced/metastatic clear cell RCC who had received ≥ 1 prior VEGFR-TKI treatment were randomized 1:1 to receive cabozantinib or everolimus. Patients were categorized by recruitment region: Europe or outside of Europe (rest of world [RoW]). Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and adverse events (AEs) were compared between regional subgroups. RESULTS: In total, there were 320 eligible patients from Europe (cabozantinib, 167; everolimus, 153) and 338 from RoW (North America, 240 patients; Asia-Pacific, 86; Latin America, 12; randomized as cabozantinib, 163; everolimus, 175). PFS and OS were longer with cabozantinib than with everolimus and similar for the Europe and RoW subgroups. For PFS, the hazard ratio (HR) for cabozantinib versus everolimus was 0.54 for the Europe subgroup (p < .001) and 0.50 for the RoW subgroup (p < .001). For OS, the HR was 0.75 for the Europe subgroup (p = .034) and 0.69 for the RoW subgroup (p = .006). ORR in the Europe subgroup was 15% for cabozantinib and 3.9% for everolimus (p < .001). For the RoW subgroup, ORR was 20% for cabozantinib and 2.9% for everolimus (p < .001). Incidence of grade 3/4 AEs were similar for the Europe (cabozantinib, 74%; everolimus, 58%) and RoW subgroups (cabozantinib, 69%; everolimus, 64%). CONCLUSION: In the METEOR trial, efficacy outcomes for patients recruited from European and non-European countries favored cabozantinib over everolimus. The efficacy and safety results for the regional subgroups were consistent with those of the overall METEOR population.


Asunto(s)
Antineoplásicos , Carcinoma de Células Renales , Neoplasias Renales , Anilidas/efectos adversos , Antineoplásicos/efectos adversos , Carcinoma de Células Renales/tratamiento farmacológico , Everolimus/efectos adversos , Humanos , Neoplasias Renales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/efectos adversos , Piridinas
2.
Cancer ; 122(24): 3856-3864, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27525386

RESUMEN

BACKGROUND: Cabozantinib significantly prolonged progression-free survival (PFS) versus a placebo in patients with progressive, metastatic medullary thyroid cancer (MTC; P < .001). An exploratory analysis of phase 3 trial data evaluated the influence of rearranged during transfection (RET) and RAS (HRAS, KRAS, and NRAS) mutations on cabozantinib clinical activity. METHODS: Patients (n = 330) were randomized to cabozantinib (140 mg/day) or a placebo. The primary endpoint was PFS. Additional outcome measures included PFS, objective response rates (ORRs), and adverse events in RET and RAS mutation subgroups. RESULTS: Among all study patients, 51.2% were RET mutation-positive (38.2% with RET M918T), 34.8% were RET mutation-unknown, and 13.9% were RET mutation-negative. Sixteen patients were RAS mutation-positive. Cabozantinib appeared to prolong PFS versus the placebo in the RET mutation-positive subgroup (hazard ratio [HR], 0.23; 95% confidence interval [CI], 0.14-0.38; P < .0001), the RET mutation-unknown subgroup (HR, 0.30; 95% CI, 0.16-0.57; P = .0001), and the RAS mutation-positive subgroup (HR, 0.15; 95% CI, 0.02-1.10; P = .0317). The RET M918T subgroup achieved the greatest observed PFS benefit from cabozantinib versus the placebo (HR, 0.15; 95% CI, 0.08-0.28; P < .0001). The ORRs for RET mutation-positive, RET mutation-negative, and RAS mutation-positive patients were 32%, 22%, and 31%, respectively. No PFS benefit was observed in patients lacking both RET and RAS mutations, although the ORR was 21%. The safety profile for all subgroups was similar to that for the overall cabozantinib arm. CONCLUSIONS: These data suggest that cabozantinib provides the greatest clinical benefit to patients with MTC who have RET M918T or RAS mutations. However, a prospective trial is needed to confirm the relation between genetic variation and the response to cabozantinib. Cancer 2016;122:3856-3864. © 2016 American Cancer Society.


Asunto(s)
Anilidas/uso terapéutico , Mutación/genética , Proteínas Proto-Oncogénicas c-ret/genética , Piridinas/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Proteínas ras/genética , Supervivencia sin Enfermedad , Método Doble Ciego , Humanos
3.
Invest New Drugs ; 33(2): 349-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25476894

RESUMEN

Background XL281 is a potent and selective inhibitor of wild-type and mutant RAF kinases with anti-tumor activity in multiple xenograft models. Mutations in KRAS or BRAF can activate the RAF/MEK/ERK pathway in human tumors and may confer sensitivity to RAF kinase inhibitors. Methods We performed a phase I study of XL281 in patients with advanced solid tumors. Patients were enrolled in successive cohorts of XL281 orally once daily in 28-day cycles. Twice daily dosing, different formulations, and the effect of food and famotidine were also studied. At the MTD expanded cohorts with defined mutations were treated. Results In total, 160 patients were treated. The MTD on the QD schedule was 150 mg. The most common toxicities were diarrhea, nausea, and fatigue. Plasma Cmax and AUC increased with dose. Famotidine resulted in lower AUC while food had no effect. Two patients had partial responses by RECIST: One with papillary thyroid cancer with NRAS mutation and one with uveal melanoma. Another nine patients had tumor decrease of >10% but did not meet RECIST criteria for PR. Matched tumors pairs from 33 patients showed evidence of RAF inhibition with significant decreases in pERK, pMEK and pAKT. Conclusions XL281 was generally well tolerated and the MTD was established at 150 mg/day. Partial responses and clinical benefit were observed in several patients. Tumor biopsies demonstrated effective target inhibition.


Asunto(s)
Antineoplásicos/farmacocinética , Bencimidazoles/farmacocinética , Carbamatos/farmacocinética , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Área Bajo la Curva , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Carbamatos/administración & dosificación , Carbamatos/efectos adversos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Antagonismo de Drogas , Famotidina/farmacología , Femenino , Interacciones Alimento-Droga , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Quinasas raf/antagonistas & inhibidores
4.
Cancer Treat Rev ; 89: 102062, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32659623

RESUMEN

Cabozantinib is an oral tyrosine kinase inhibitor (TKI) approved for the treatment of patients with advanced renal cell carcinoma (RCC) at a dose of 60 mg/day. As with other TKIs, cabozantinib is associated with high interpatient variability in drug clearance and exposure that can significantly impact safety and tolerability across a patient population. To optimize cabozantinib exposure (maintaining efficacy and tolerability) for the individual, patients may require treatment interruption with dose reduction (40 mg/day and then 20 mg/day). In the pivotal Phase 3 METEOR trial, cabozantinib significantly improved overall survival, progression-free survival and the objective response rate compared with everolimus in patients with advanced RCC who had received previous treatment with a VEGFR TKI. Dose reductions were common for patients receiving cabozantinib (60%) but effective as only 9% discontinued treatment due to adverse events (AEs). In this review, we discuss pharmacometric analyses that evaluated the impact of cabozantinib dose on efficacy and safety outcomes during the METEOR study. Exposure-response models demonstrate that the risk of experiencing adverse events and dose reduction is increased in patients with low cabozantinib clearance versus typical clearance and decreased in patients with high clearance. Dose reduction of cabozantinib to manage AEs is predicted to have minimal impact on efficacy as AEs are more likely to occur in patients with low clearance and higher exposure to cabozantinib. These analyses further support a dose modification strategy to optimize cabozantinib exposure for individual patients.


Asunto(s)
Anilidas/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Piridinas/administración & dosificación , Anilidas/efectos adversos , Anilidas/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Carcinoma de Células Renales/metabolismo , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Renales/metabolismo , Modelos Estadísticos , Supervivencia sin Progresión , Piridinas/efectos adversos , Piridinas/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/efectos adversos , Proteínas Tirosina Quinasas Receptoras/farmacocinética
5.
J Comp Neurol ; 495(2): 202-12, 2006 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-16435287

RESUMEN

During embryonic development, complex events, such as cellular proliferation, differentiation, survival, and guidance of axons, are orchestrated and regulated by a variety of extracellular signals. Receptor tyrosine kinases mediate many of these events, with several playing critical roles in neuronal survival and axonal guidance. It is evident that not all the receptor tyrosine kinases that play key roles in regulating neuronal development have been identified. In this study, we have characterized the spatial-temporal expression profile of a recently identified receptor tyrosine kinase, anaplastic lymphoma kinase (ALK), in embryonic chick by means of whole-mount in situ hybridization in conjunction with immunohistochemistry. Our findings reveal that Alk is expressed in sympathetic and dorsal root ganglia as early as stage 19. In addition, mRNA is expressed from stage 23/24 (E4) to stage 39 (E13) in discrete motor neuron subsets of chick spinal cord along with a select group of muscles that are innervated by one of these particular motor neuron clusters. Expression within the spinal cord is coincident with the onset and duration of motor neuron programmed cell death and during the period of musculature innervation and synapse formation. Hence, the data presented here identify ALK as a novel candidate receptor for regulating critical events in the development of neurons in both the central and the peripheral nervous systems.


Asunto(s)
Tipificación del Cuerpo/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas Motoras/metabolismo , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Quinasa de Linfoma Anaplásico , Animales , Embrión de Pollo , Desnervación/métodos , Extremidades/inervación , Expresión Génica/fisiología , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Modelos Biológicos , Neuronas Motoras/clasificación , Sistema Nervioso Periférico/embriología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas Receptoras , Médula Espinal/embriología , Médula Espinal/metabolismo
6.
J Biomol Screen ; 9(3): 216-22, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15140383

RESUMEN

To facilitate the characterization of proteins that negatively regulate tumor cell proliferation in vitro, the authors have implemented a high-throughput functional assay that measures S-phase progression of tumor cell lines. For 2 tumor cell lines-human melanoma A375 and human lung carcinoma A549-conditions were established using the cyclin-dependent kinase inhibitor, p27kip; the tumor suppressor p53, a kinase-inactive allele of the cell cycle-regulated serine/threonine kinase Aurora2; and the G1/S drug block, aphidicolin. For screening purposes, gene libraries were delivered by adenoviral infection. Cells were fixed and labeled by immunocytochemistry, and an automated image acquisition and analysis package on a Cellomics ArrayScanII was used to quantify the effects of these treatments on cell proliferation. The assay can be used to identify novel proteins involved in proliferation and serves as a more robust, reproducible, and sensitive alternative to enzyme-linked immunosorbent assay (ELISA)-based technologies.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Inmunohistoquímica/métodos , Proteínas de Neoplasias/análisis , Neoplasias/patología , Automatización , Bromodesoxiuridina/metabolismo , Carcinoma/metabolismo , Carcinoma/patología , División Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Fluorescencia , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Melanoma/metabolismo , Melanoma/patología , Neoplasias/metabolismo , Sensibilidad y Especificidad , Células Tumorales Cultivadas
7.
J Biomol Screen ; 9(8): 704-11, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15634797

RESUMEN

With the sequence of the human genome at hand, target discovery strategies are needed that can rapidly identify novel gene products involved in human disease pathways. In this article, the authors describe a cell-based, high-throughput assay that can identify gene products capable of modulating the vascular endothelial growth factor (VEGF) and tumor necrosis factor alpha (TNFalpha) signaling pathways in human endothelial cells. The assay uses real-time PCR technology to measure downstream reporter mRNA transcripts induced upon cytokine stimulation in a 96-well plate format and has been adapted for use with recombinant adenoviruses. The authors specifically demonstrate modulation of cytokine-driven reporter transcripts using drug inhibitors and through adenoviral-mediated expression of known signaling intermediates of the respective pathways. In addition, they have used an arrayed library of 350 recombinant adenoviruses to screen for novel modulators of the VEGF and TNFalpha pathways. The high-throughput screening capacity and sensitivity of this system make it a useful tool for new drug target identification.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Endotelio Vascular/efectos de los fármacos , Genómica/métodos , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Adenoviridae/genética , Bioensayo , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Biblioteca de Genes , Técnicas de Transferencia de Gen , Genes Reporteros , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Tromboplastina/genética , Tromboplastina/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
8.
Leuk Res ; 38(3): 316-22, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24374145

RESUMEN

This phase I study evaluated selective JAK2 inhibitor XL019 in 30 patients with myelofibrosis. The initial dose cohorts were 100, 200, and 300 mg orally on days 1-21 of a 28-day cycle. Central and/or peripheral neurotoxicity developed in all patients. Subsequently, patients were treated on lower doses; neurotoxicity was again observed, leading to study termination. Peripheral neuropathy resolved in 50%, and central neurotoxicity in all patients within months after therapy cessation. Myelosuppression was minimal. The terminal half-life of XL019 was approximately 21 h, with steady state reached by Day 8. International Working Group defined responses were seen in three (10%) patients.


Asunto(s)
Antineoplásicos/efectos adversos , Terminación Anticipada de los Ensayos Clínicos , Janus Quinasa 2/antagonistas & inhibidores , Síndromes de Neurotoxicidad/etiología , Mielofibrosis Primaria/tratamiento farmacológico , Prolina/análogos & derivados , Inhibidores de Proteínas Quinasas/efectos adversos , Pirimidinas/efectos adversos , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Esquema de Medicación , Monitoreo de Drogas , Femenino , Humanos , Janus Quinasa 2/genética , Masculino , Persona de Mediana Edad , Síndromes de Neurotoxicidad/diagnóstico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Prolina/administración & dosificación , Prolina/efectos adversos , Prolina/farmacocinética , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética
9.
Cell Cycle ; 6(1): 104-10, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17245119

RESUMEN

Chk1 and Chk2 kinases are critically involved in modulating DNA damage checkpoints. In particular, Chk1, a key activator of the S-phase DNA damage response, may be involved in resistance to genotoxic therapies that target DNA synthesis. We studied the in vitro and in vivo effects of EXEL-9844 (XL844), a potent, orally available, and specific inhibitor of Chk1 and Chk2, in combination with gemcitabine. In clonogenic assays using multiple cell lines in vitro, EXEL-9844 had only minor effects as a single agent but substantially enhanced gemcitabine-induced cell killing. Correspondingly, in PANC-1 cells, EXEL-9844 increased gemcitabine-induced H2AX phosphorylation, blocked Cdc25A phosphorylation, and induced premature mitotic entry. In a PANC-1 xenograft model, EXEL-9844 significantly enhanced gemcitabine antitumor activity but had limited effect as a single agent. Together, these data show that cell cycle checkpoint inhibitors may have significant clinical utility in potentiating the activity of gemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Desoxicitidina/análogos & derivados , Genes cdc/efectos de los fármacos , Fase S/efectos de los fármacos , Animales , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Genes cdc/fisiología , Ratones , Ratones Desnudos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Gemcitabina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA