Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Ann Oncol ; 33(12): 1269-1283, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36089135

RESUMEN

BACKGROUND: Targeted therapies have transformed clinical management of advanced biliary tract cancer (BTC). Cell-free DNA (cfDNA) analysis is an attractive approach for cancer genomic profiling that overcomes many limitations of traditional tissue-based analysis. We examined cfDNA as a tool to inform clinical management of patients with advanced BTC and generate novel insights into BTC tumor biology. PATIENTS AND METHODS: We analyzed next-generation sequencing data of 2068 cfDNA samples from 1671 patients with advanced BTC generated with Guardant360. We carried out clinical annotation on a multi-institutional subset (n = 225) to assess intra-patient cfDNA-tumor concordance and the association of cfDNA variant allele fraction (VAF) with clinical outcomes. RESULTS: Genetic alterations were detected in cfDNA in 84% of patients, with targetable alterations detected in 44% of patients. Fibroblast growth factor receptor 2 (FGFR2) fusions, isocitrate dehydrogenase 1 (IDH1) mutations, and BRAF V600E were clonal in the majority of cases, affirming these targetable alterations as early driver events in BTC. Concordance between cfDNA and tissue for mutation detection was high for IDH1 mutations (87%) and BRAF V600E (100%), and low for FGFR2 fusions (18%). cfDNA analysis uncovered novel putative mechanisms of resistance to targeted therapies, including mutation of the cysteine residue (FGFR2 C492F) to which covalent FGFR inhibitors bind. High pre-treatment cfDNA VAF was associated with poor prognosis and shorter response to chemotherapy and targeted therapy. Finally, we report the frequency of promising targets in advanced BTC currently under investigation in other advanced solid tumors, including KRAS G12C (1.0%), KRAS G12D (5.1%), PIK3CA mutations (6.8%), and ERBB2 amplifications (4.9%). CONCLUSIONS: These findings from the largest and most comprehensive study to date of cfDNA from patients with advanced BTC highlight the utility of cfDNA analysis in current management of this disease. Characterization of oncogenic drivers and mechanisms of therapeutic resistance in this study will inform drug development efforts to reduce mortality for patients with BTC.


Asunto(s)
Neoplasias de los Conductos Biliares , Neoplasias del Sistema Biliar , Ácidos Nucleicos Libres de Células , Humanos , Ácidos Nucleicos Libres de Células/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias de los Conductos Biliares/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación , Neoplasias del Sistema Biliar/tratamiento farmacológico , Neoplasias del Sistema Biliar/genética , Neoplasias del Sistema Biliar/patología
2.
Sci Adv ; 5(5): eaau5240, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31086813

RESUMEN

The Wnt/ß-catenin (ß-cat) pathway plays a critical role in cancer. Using hydrocarbon-stapled peptide technologies, we aim to develop potent, selective inhibitors targeting this pathway by disrupting the interaction of ß-cat with its coactivators B-cell lymphoma 9 (BCL9) and B-cell lymphoma 9-like (B9L). We identified a set of peptides, including hsBCL9CT-24, that robustly inhibits the activity of ß-cat and suppresses cancer cell growth. In animal models, these peptides exhibit potent anti-tumor effects, favorable pharmacokinetic profiles, and minimal toxicities. Markedly, these peptides promote intratumoral infiltration of cytotoxic T cells by reducing regulatory T cells (Treg) and increasing dendritic cells (DCs), therefore sensitizing cancer cells to PD-1 inhibitors. Given the strong correlation between Treg infiltration and APC mutation in colorectal cancers, it indicates our peptides can reactivate anti-cancer immune response suppressed by the oncogenic Wnt pathway. In summary, we report a promising strategy for cancer therapy by pharmacological inhibition of the Wnt/ß-cat signaling.


Asunto(s)
Linfocitos T Reguladores/inmunología , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Antineoplásicos Inmunológicos/metabolismo , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL20/antagonistas & inhibidores , Quimiocina CCL20/metabolismo , Quimiocina CCL22/antagonistas & inhibidores , Quimiocina CCL22/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Péptidos/metabolismo , Péptidos/farmacología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Trasplante Heterólogo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/antagonistas & inhibidores
3.
Ann Oncol ; 28(12): 2932-2942, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28950290

RESUMEN

BACKGROUND: Ramucirumab, the human immunoglobulin G1 monoclonal antibody receptor antagonist of vascular endothelial growth factor receptor 2, has been approved for treating gastric/gastroesophageal junction, non-small-cell lung, and metastatic colorectal cancers. With the completion of six global, randomized, double-blind, placebo-controlled, phase III trials across multiple tumor types, an opportunity now exists to further establish the safety parameters of ramucirumab across a large patient population. MATERIALS AND METHODS: An individual patient meta-analysis across the six completed phase III trials was conducted and the relative risk (RR) and associated 95% confidence intervals (CIs) were derived using fixed-effects or mixed-effects models for all-grade and high-grade adverse events (AEs) possibly related to vascular endothelial growth factor pathway inhibition. The number needed to harm was also calculable due to the placebo-controlled nature of all six registration standard trials. RESULTS: A total of 4996 treated patients (N = 2748 in the ramucirumab arm and N = 2248 in the control, placebo arm) were included in this meta-analysis. Arterial thromboembolic events [ATE; all-grade, RR: 0.8, 95% CI 0.5-1.3; high-grade (grade ≥3), RR: 0.9, 95% CI 0.5-1.7], venous thromboembolic events (VTE; all-grade, RR: 0.7, 95% CI 0.5-1.1; high-grade, RR: 0.7, 95% CI 0.4-1.2), high-grade bleeding (RR: 1.1, 95% CI 0.8-1.5), and high-grade gastrointestinal (GI) bleeding (RR: 1.1, 95% CI 0.7-1.7) did not demonstrate a definite increased risk with ramucirumab. A higher percentage of hypertension, proteinuria, low-grade (grade 1-2) bleeding, GI perforation, infusion-related reaction, and wound-healing complications were observed in the ramucirumab arm compared with the control arm. CONCLUSIONS: Ramucirumab may be distinct among antiangiogenic agents in terms of ATE, VTE, high-grade bleeding, or high-grade GI bleeding by showing no clear evidence for an increased risk of these AEs in this meta-analysis of a large and diverse patient population. Ramucirumab is consistent with other angiogenic inhibitors in the risk of developing certain AEs. Clinical Trial Numbers: NCT00917384 (REGARD), NCT01170663 (RAINBOW), NCT01168973 (REVEL), NCT01183780 (RAISE), NCT01140347 (REACH), and NCT00703326 (ROSE).


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/inmunología , Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ensayos Clínicos Fase III como Asunto , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Medición de Riesgo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Ramucirumab
4.
Bull Entomol Res ; 106(6): 809-817, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27443747

RESUMEN

Extreme high-temperature events are the key factor to determine population dynamics of the rice leaf folder, Cnaphalocrocis medinalis (Guenée), in summer. Although we know that adult of this insect can migrate to avoid heat stress, the behavioral response of larva to high temperature is still unclear. Therefore, impacts of high temperature on behavioral traits of C. medinalis including host choice, settling and folding leaf were observed. The results revealed that these behavioral traits were clearly influenced by high temperature. The larvae preferred maize leaves rather than rice and wheat at normal temperature of 27°C, but larvae experienced a higher temperature of 37 or 40°C for 4 h preferred rice leaves rather than maize and wheat. Capacity of young larvae to find host leaves or settle on the upper surface of leaves significantly reduced when they were treated by high temperature. High temperature of 40°C reduced the leaf-folding capacity of the third instar larvae, but no effects were observed on the fourth and fifth instar larvae. Short-term heat acclimation could not improve the capacity of the third instar larvae to make leaf fold under 40°C.


Asunto(s)
Conducta Animal , Mariposas Nocturnas/fisiología , Oryza , Estrés Fisiológico , Animales , Conducta Alimentaria , Respuesta al Choque Térmico , Larva/fisiología , Temperatura , Zea mays
5.
Braz. j. med. biol. res ; 47(9): 738-745, 09/2014. graf
Artículo en Inglés | LILACS | ID: lil-719313

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is one of the most potent angiogenic growth factors. It improves angiogenesis and tissue perfusion in ischemic skeletal muscle. In the present study, we tested the hypothesis that ischemic postconditioning is effective for salvaging ischemic skeletal muscle resulting from limb ischemia-reperfusion injury, and that the mechanism involves expression of HIF-1α. Wistar rats were randomly divided into three groups (n=36 each): sham-operated (group S), hindlimb ischemia-reperfusion (group IR), and ischemic postconditioning (group IPO). Each group was divided into subgroups (n=6) according to reperfusion time: immediate (0 h, T0), 1 h (T1), 3 h (T3), 6 h (T6), 12 h (T12), and 24 h (T24). In the IPO group, three cycles of 30-s reperfusion and 30-s femoral aortic reocclusion were carried out before reperfusion. At all reperfusion times (T0-T24), serum creatine kinase (CK) and lactate dehydrogenase (LDH) activities, as well as interleukin (IL)-6, IL-10, and tumor necrosis factor-α (TNF-α) concentrations, were measured in rats after they were killed. Histological and immunohistochemical methods were used to assess the skeletal muscle damage and HIF-1α expression in skeletal muscle ischemia. In groups IR and IPO, serum LDH and CK activities and TNF-α, IL-6, and IL-10 concentrations were all significantly increased compared to group S, and HIF-1α expression was up-regulated (P<0.05 or P<0.01). In group IPO, serum LDH and CK activities and TNF-α and IL-6 concentrations were significantly decreased, IL-10 concentration was increased, HlF-1α expression was down-regulated (P<0.05 or P<0.01), and the pathological changes were reduced compared to group IR. The present study suggests that ischemic postconditioning can reduce skeletal muscle damage caused by limb ischemia-reperfusion and that its mechanisms may be related to the involvement of HlF-1α in the limb ischemia-reperfusion injury-triggered inflammatory response.


Asunto(s)
Animales , Masculino , Extremidades/irrigación sanguínea , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Poscondicionamiento Isquémico , Músculo Esquelético/irrigación sanguínea , Daño por Reperfusión/prevención & control , Western Blotting , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Inmunohistoquímica , /sangre , /sangre , L-Lactato Deshidrogenasa/metabolismo , Músculo Esquelético/lesiones , Distribución Aleatoria , Ratas Wistar , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre , Regulación hacia Arriba
6.
Braz J Med Biol Res ; 47(9): 738-45, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25075575

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is one of the most potent angiogenic growth factors. It improves angiogenesis and tissue perfusion in ischemic skeletal muscle. In the present study, we tested the hypothesis that ischemic postconditioning is effective for salvaging ischemic skeletal muscle resulting from limb ischemia-reperfusion injury, and that the mechanism involves expression of HIF-1α. Wistar rats were randomly divided into three groups (n=36 each): sham-operated (group S), hindlimb ischemia-reperfusion (group IR), and ischemic postconditioning (group IPO). Each group was divided into subgroups (n=6) according to reperfusion time: immediate (0 h, T0), 1 h (T1), 3 h (T3), 6 h (T6), 12 h (T12), and 24 h (T24). In the IPO group, three cycles of 30-s reperfusion and 30-s femoral aortic reocclusion were carried out before reperfusion. At all reperfusion times (T0-T24), serum creatine kinase (CK) and lactate dehydrogenase (LDH) activities, as well as interleukin (IL)-6, IL-10, and tumor necrosis factor-α (TNF-α) concentrations, were measured in rats after they were killed. Histological and immunohistochemical methods were used to assess the skeletal muscle damage and HIF-1α expression in skeletal muscle ischemia. In groups IR and IPO, serum LDH and CK activities and TNF-α, IL-6, and IL-10 concentrations were all significantly increased compared to group S, and HIF-1α expression was up-regulated (P<0.05 or P<0.01). In group IPO, serum LDH and CK activities and TNF-α and IL-6 concentrations were significantly decreased, IL-10 concentration was increased, HlF-1α expression was down-regulated (P<0.05 or P<0.01), and the pathological changes were reduced compared to group IR. The present study suggests that ischemic postconditioning can reduce skeletal muscle damage caused by limb ischemia-reperfusion and that its mechanisms may be related to the involvement of HlF-1α in the limb ischemia-reperfusion injury-triggered inflammatory response.


Asunto(s)
Extremidades/irrigación sanguínea , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Poscondicionamiento Isquémico , Músculo Esquelético/irrigación sanguínea , Daño por Reperfusión/prevención & control , Animales , Western Blotting , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Inmunohistoquímica , Interleucina-10/sangre , Interleucina-6/sangre , L-Lactato Deshidrogenasa/metabolismo , Masculino , Músculo Esquelético/lesiones , Distribución Aleatoria , Ratas Wistar , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre , Regulación hacia Arriba
7.
Br J Cancer ; 111(3): 430-6, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-24960403

RESUMEN

BACKGROUND: Current data suggest that platinum-based combination therapy is the standard first-line treatment for biliary tract cancer. EGFR inhibition has proven beneficial across a number of gastrointestinal malignancies; and has shown specific advantages among KRAS wild-type genetic subtypes of colon cancer. We report the combination of panitumumab with gemcitabine (GEM) and oxaliplatin (OX) as first-line therapy for KRAS wild-type biliary tract cancer. METHODS: Patients with histologically confirmed, previously untreated, unresectable or metastatic KRAS wild-type biliary tract or gallbladder adenocarcinoma with ECOG performance status 0-2 were treated with panitumumab 6 mg kg(-1), GEM 1000 mg m(-2) (10 mg m(-2) min(-1)) and OX 85 mg m(-2) on days 1 and 15 of each 28-day cycle. The primary objective was to determine the objective response rate by RECIST criteria v.1.1. Secondary objectives were to evaluate toxicity, progression-free survival (PFS), and overall survival. RESULTS: Thirty-one patients received at least one cycle of treatment across three institutions, 28 had measurable disease. Response rate was 45% and disease control rate was 90%. Median PFS was 10.6 months (95% CI 5-24 months) and median overall survival 20.3 months (95% CI 9-25 months). The most common grade 3/4 adverse events were anaemia 26%, leukopenia 23%, fatigue 23%, neuropathy 16% and rash 10%. CONCLUSIONS: The combination of gemcitabine, oxaliplatin and panitumumab in KRAS wild type metastatic biliary tract cancer showed encouraging efficacy, additional efforts of genetic stratification and targeted therapy is warranted in biliary tract cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Sistema Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Neoplasias del Sistema Biliar/mortalidad , Neoplasias del Sistema Biliar/patología , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Supervivencia sin Enfermedad , Femenino , Neoplasias de la Vesícula Biliar/mortalidad , Neoplasias de la Vesícula Biliar/patología , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Panitumumab , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Resultado del Tratamiento , Proteínas ras/genética , Gemcitabina
8.
Ann Oncol ; 25(1): 121-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24356623

RESUMEN

BACKGROUND: To determine the maximal tolerated dose of erlotinib when added to 5-fluorouracil (5-FU) chemoradiation and bevacizumab and safety and efficacy of this combination in patients with locally advanced rectal cancer. PATIENTS AND METHODS: Patients with Magnetic resonance imaging (MRI) or ultrasound defined T3 or T4 adenocarcinoma of the rectum and without evidence of metastatic disease were enrolled. Patients received infusional 5-FU 225 mg/M2/day continuously, along with bevacizumab 5 mg/kg days 14, 1, 15 and 29. Standard radiotherapy was administered to 50.4 Gy in 28 fractions. Erlotinib started at a dose of 50 mg orally daily and advanced by 50 mg increments in the subsequent cohort. Open total mesorectal excision was carried out 6-9 weeks following the completion of chemoradiation. RESULTS: Thirty-two patients received one of three dose levels of erlotinib. Erlotinib dose level of 100 mg was determined to be the maximally tolerated dose. Thirty-one patients underwent resection of the primary tumor, one refused resection. Twenty-seven patients completed study therapy, all of whom underwent resection. At least one grade 3-4 toxicity occurred in 46.9% of patients. Grade 3-4 diarrhea occurred in 18.8%. The pathologic complete response (pCR) for all patients completing study therapy was 33%. With a median follow-up of 2.9 years, there are no documented local recurrences. Disease-free survival at 3 years is 75.5% (confidence interval: 55.1-87.6%). CONCLUSIONS: Erlotinib added to infusional 5-FU, bevacizumab and radiation in patients with locally advanced rectal cancer is relatively well tolerated and associated with an encouraging pCR.


Asunto(s)
Adenocarcinoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Recto/terapia , Anticuerpos Monoclonales Humanizados/administración & dosificación , Bevacizumab , Quimioradioterapia , Quimioterapia Adyuvante , Supervivencia sin Enfermedad , Clorhidrato de Erlotinib , Femenino , Fluorouracilo/administración & dosificación , Humanos , Masculino , Terapia Neoadyuvante , Quinazolinas/administración & dosificación , Resultado del Tratamiento
9.
Br J Cancer ; 108(7): 1393-401, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23511559

RESUMEN

BACKGROUND: This phase I, dose-finding study determined the maximum tolerated dose (MTD), safety, and pharmacokinetics of sunitinib plus gemcitabine in patients with advanced solid tumours. METHODS: Two schedules with sunitinib (25-50 mg per day) and IV gemcitabine (750-1250 mg m(-2)) in escalating doses were studied. First, patients received sunitinib on a 4-weeks-on-2-weeks-off schedule (Schedule 4/2) plus gemcitabine on days 1, 8, 22, and 29. Second, patients received sunitinib on a 2-weeks-on-1-week-off schedule (Schedule 2/1) plus gemcitabine on days 1 and 8. The primary endpoint was determination of MTD and tolerability. RESULTS: Forty-four patients received the combination (Schedule 4/2, n=8; Schedule 2/1, n=36). With no dose-limiting toxicities (DLTs) at maximum dose levels on Schedule 2/1, MTD was not reached. Grade 4 treatment-related AEs and laboratory abnormalities included cerebrovascular accident, hypertension, and pulmonary embolism (n=1 each), and neutropenia (n=3), thrombocytopenia and increased uric acid (both n=2), and lymphopenia (n=1). There were no clinically significant drug-drug interactions. Antitumor activity occurred across dose levels and tumour types. In poor-risk and/or high-grade renal cell carcinoma patients (n=12), 5 had partial responses and 7 stable disease ≥ 6 weeks. CONCLUSION: Sunitinib plus gemcitabine on Schedule 2/1 with growth factor support was well tolerated and safely administered at maximum doses of each drug, without significant drug-drug interactions.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Indoles/administración & dosificación , Indoles/efectos adversos , Indoles/farmacocinética , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/patología , Pirroles/administración & dosificación , Pirroles/efectos adversos , Pirroles/farmacocinética , Sunitinib , Gemcitabina
10.
Ann Oncol ; 22(6): 1367-1373, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21217058

RESUMEN

BACKGROUND: Epidermal growth factor receptor (EGFR) is overexpressed in a significant proportion of esophageal and gastric carcinomas. Although previous studies have examined tyrosine kinase inhibitors of EGFR, there remains limited data regarding the role of EGFR-directed monoclonal antibody therapy in these malignancies. We carried out a multi-institutional phase II study of cetuximab, a monoclonal antibody against EGFR, in patients with unresectable or metastatic esophageal or gastric adenocarcinoma. PATIENTS AND METHODS: Thirty-five patients with previously treated metastatic esophageal or gastric adenocarcinoma were treated with weekly cetuximab, at an initial dose of 400 mg/m(2) followed by weekly infusions at 250 mg/m(2). Patients were followed for toxicity, treatment response, and survival. RESULTS: Treatment with cetuximab was well tolerated; no patients were taken off study due to drug-related adverse events. One (3%) partial treatment response was noted. Two (6%) patients had stable disease after 2 months of treatment. Median progression-free survival and overall survival were 1.6 and 3.1 months, respectively. CONCLUSION: Although well tolerated, cetuximab administered as a single agent had minimal clinical activity in patients with metastatic esophageal and gastric adenocarcinoma. Ongoing studies of EGFR inhibitors in combination with other agents may define a role for these agents in the treatment of esophageal and gastric cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Antineoplásicos/efectos adversos , Cetuximab , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
11.
Oncogene ; 29(36): 4989-5005, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20639898

RESUMEN

Hepatocellular carcinoma (HCC) is a highly prevalent, treatment-resistant malignancy with a multifaceted molecular pathogenesis. Current evidence indicates that during hepatocarcinogenesis, two main pathogenic mechanisms prevail: (1) cirrhosis associated with hepatic regeneration after tissue damage caused by hepatitis infection, toxins (for example, alcohol or aflatoxin) or metabolic influences, and (2) mutations occurring in single or multiple oncogenes or tumor suppressor genes. Both mechanisms have been linked with alterations in several important cellular signaling pathways. These pathways are of interest from a therapeutic perspective, because targeting them may help to reverse, delay or prevent tumorigenesis. In this review, we explore some of the major pathways implicated in HCC. These include the RAF/MEK/ERK pathway, phosphatidylinositol-3 kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, WNT/beta-catenin pathway, insulin-like growth factor pathway, hepatocyte growth factor/c-MET pathway and growth factor-regulated angiogenic signaling. We focus on the role of these pathways in hepatocarcinogenesis, how they are altered, and the consequences of these abnormalities. In addition, we also review the latest preclinical and clinical data on the rationally designed targeted agents that are now being directed against these pathways, with early evidence of success.


Asunto(s)
Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/etnología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/terapia , Transducción de Señal/fisiología , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Receptores ErbB/genética , Receptores ErbB/fisiología , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Sistema de Señalización de MAP Quinasas/fisiología , Modelos Biológicos , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Transducción de Señal/genética , Somatomedinas/genética , Somatomedinas/fisiología
12.
Ann Oncol ; 20(3): 475-80, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19139178

RESUMEN

BACKGROUND: Recent studies have examined the addition of docetaxel to fluorouracil and cisplatin in advanced esophagogastric cancer. PATIENTS AND METHODS: We carried out a phase I dose-escalation study of weekly docetaxel, cisplatin, and irinotecan (TPC), given on days 1 and 8 every 3 weeks, in patients with chemonaive solid tumors. Subsequently, we completed a multiinstitutional phase II study of TPC in patients with previously untreated, metastatic esophagogastric cancer. RESULTS: Thirty-nine patients were enrolled in the phase I trial; a weekly schedule of TPC was well tolerated. On that basis, docetaxel 30 mg/m(2), cisplatin 25 mg/m(2), and irinotecan 65 mg/m(2) were selected for the phase II trial, where in the first 18 patients irinotecan 65 mg/m(2) caused too much diarrhea and was reduced to 50 mg/m(2). Among 56 eligible patients with previously untreated, metastatic esophagogastric cancer enrolled in the phase II trial, three complete and 27 partial responses were observed (overall response rate=54%), and 15 patients (30%) had stable disease. Median progression-free survival was 7.1 months, and median survival was 11.9 months. At the final irinotecan dose of 50 mg/m(2), grade 3 or higher toxicity included diarrhea (26%), neutropenia (21%), nausea (18%), fatigue (16%), anorexia (13%), and thrombosis/embolism (13%). CONCLUSIONS: Weekly TPC is an active and well-tolerated regimen for patients with esophagogastric cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Cisplatino/administración & dosificación , Docetaxel , Neoplasias Esofágicas/patología , Femenino , Humanos , Irinotecán , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias Gástricas/patología , Taxoides/administración & dosificación
13.
Ann Oncol ; 18(7): 1185-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17483115

RESUMEN

BACKGROUND: Targeting the epidermal growth factor receptor and angiogenesis have proven useful strategies against metastatic colorectal cancer. The benefit of combining inhibitors of both pathways is unknown. PATIENTS AND METHODS: Patients with previously untreated metastatic colorectal cancer were enrolled in a phase II trial of infusional 5-fluorouracil, leucovorin, oxaliplatin (FOLFOX), bevacizumab and erlotinib. The primary end point was progression-free survival. RESULTS: Thirty-five patients were enrolled and all came off trial for reasons other than progression; 18 (51%) had protocol-defined adverse events requiring removal, nine (26%) withdrew consent due to toxicity, six pursued surgery or localized therapies and two requested a treatment holiday. Principal toxic effects included rash, neuropathy and diarrhea. Seven patients came off trial before first restaging. By intention-to-treat analysis, one patient had a confirmed complete response, 10 had confirmed partial responses and one had an unconfirmed partial response (response rate = 34%). One patient had progressive disease at time of withdrawal from the trial, thus progression-free survival could not be calculated. CONCLUSION: The combination of FOLFOX, bevacizumab and erlotinib led to higher than expected early withdrawal due to toxicity, limiting conclusions regarding efficacy. These findings raise concern regarding the tolerability of adding more agents to already complex combination regimens for metastatic colorectal cancer.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias Colorrectales/tratamiento farmacológico , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Bevacizumab , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Clorhidrato de Erlotinib , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Humanos , Estimación de Kaplan-Meier , Leucovorina/administración & dosificación , Leucovorina/efectos adversos , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/efectos adversos , Quinazolinas/administración & dosificación , Quinazolinas/efectos adversos
14.
Cancer Chemother Pharmacol ; 60(2): 203-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17091249

RESUMEN

PURPOSE: MAC-321 is a novel taxane that has demonstrated exceptional activity in human xenograft models when administered intravenously and orally. Preclinical studies of MAC-321 have shown antitumor activity in MDR-expressing and paclitaxel-resistant tumors. This phase I dose escalation study was performed to determine the safety, tolerability, and pharmacokinetic profile of orally administered MAC-321 given once every 21 days. Preliminary antitumor activity of MAC-321 was also examined. METHODS: Key eligibility criteria included adult subjects with refractory solid tumors or solid tumors for which conventional therapy was unsuitable or did not exist, good performance status (ECOG ( 2), and adequate hematologic, hepatic, and renal functions. Plasma pharmacokinetic (PK) sampling was performed during the first cycle of therapy. RESULTS: Five dose levels of MAC-321 ranging from 25 to 75 mg/m(2) were evaluated in 18 subjects (four women and 14 men). MAC-321 was well tolerated at the first three dose levels (25, 37, 50 mg/m(2)). Two subjects developed dose-limiting toxicities (DLTs) at 75 mg/m(2); one subject with grade 3 and one subject with grade 4 neutropenia with fever. Three subjects treated at an intermediate dose level of 60 mg/m(2) had no DLTs. However, the study was terminated prior to completion of the maximal tolerated dose cohort after subjects treated with intravenous MAC-321 in a concurrent study experienced life-threatening toxicities. Other common toxicities included grades 1-2 fatigue and grades 1-2 diarrhea. There was substantial interpatient variability in the PK parameters. MAC-321 was rapidly absorbed with a mean C (max) value of less than 1 h. Mean C (max) and AUC values generally increased in a dose-related manner. The median terminal phase elimination half-life was 45 h (range 20-228 h). Disease stabilization was seen in four subjects with the following tumors: mesothelioma (14 cycles), chondrosarcoma (12 cycles), small cell carcinoma (10 cycles), and prostate carcinoma (6 cycles). CONCLUSIONS: MAC-321 can be safely administered orally once every 21 days up to a dose of 60 mg/m(2). The major DLT was neutropenic fever. Four subjects had disease stabilization.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Paclitaxel/análogos & derivados , Administración Oral , Adulto , Anciano , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacocinética , Área Bajo la Curva , Disponibilidad Biológica , Relación Dosis-Respuesta a Droga , Femenino , Fiebre/inducido químicamente , Semivida , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/patología , Neutropenia/inducido químicamente , Paclitaxel/efectos adversos , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Resultado del Tratamiento
17.
J Biol Chem ; 271(19): 11347-55, 1996 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-8626688

RESUMEN

To determine the role of Ras-dependent signaling pathways in adipocyte function, we created transgenic mice that overexpress Ha-ras in adipocytes using the aP2 fatty acid-binding protein promoter/enhancer ligated to the human genomic ras sequence. ras mRNA was increased 8-17-fold and Ras protein 4-5-fold in white and brown fat, with no overexpression in other tissues. The subcellular distribution of overexpressed Ras paralleled that of endogenous Ras. [U-14C]Glucose uptake into isolated adipocytes was increased approximately 2-fold in the absence of insulin, and the ED50 for insulin was reduced 70%, with minimal effect on maximally stimulated glucose transport. Expression of Glut4 protein was unaltered in transgenic adipocytes, but photoaffinity labeling of transporters in intact cells with [3H]2-N-[4-(1-azi-Z,Z,Z-trifluoroethyl)benzoyl]-1,3-bis-(D-mann os-4- yloxy)-2-propylamine revealed 1.7-2.6-fold more cell-surface Glut 4 in the absence of insulin and at half-maximal insulin concentration (0.3 nM) compared with nontransgenic adipocytes. With maximal insulin concentration (80 nM), cell-surface Glut4 in nontransgenic and transgenic adipocytes was similar. Glut1 expression and basal cell-surface Glut1 were increased 2-2.9-fold in adipocytes of transgenic mice. However, Glut1 was much less abundant than Glut4, making its contribution to transport negligible. These in vitro changes were accompanied by in vivo alterations including increased glucose tolerance, decreased plasma insulin levels, and decreased adipose mass. We conclude that ras overexpression in adipocytes leads to a partial translocation of Glut4 in the absence of insulin and enhanced Glut4 translocation at physiological insulin concentration, but no effect with maximally stimulating insulin concentrations.


Asunto(s)
Tejido Adiposo/metabolismo , Expresión Génica , Genes ras , Glucosa/metabolismo , Insulina/farmacología , Proteínas Musculares , Proteínas ras/biosíntesis , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Glucemia/metabolismo , Células Cultivadas , Femenino , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 1 , Transportador de Glucosa de Tipo 4 , Humanos , Insulina/sangre , Cinética , Masculino , Ratones , Ratones Transgénicos , Proteínas de Transporte de Monosacáridos/biosíntesis , Especificidad de Órganos , Caracteres Sexuales , Proteínas ras/genética
18.
Biochem Biophys Res Commun ; 205(3): 1875-82, 1994 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-7811277

RESUMEN

Two distinct cDNAs encoding small GTP-binding proteins were isolated from a human fetal skeletal muscle cDNA library. Nucleotide sequence analysis revealed that one clone encodes a protein of 218 amino acids, which shares highest amino acid identities (over 90%) with the members of YPT3 GTP-binding proteins subfamily and was termed H-YPT3; and the second clone encodes a protein of 201 amino acids, which exhibits strongest homology to rab1A, rab1B, and YPT1 at its N-terminus but diverges notably from these rab family members at the C-terminus. The second cDNA may represents a novel member of the small GTP-binding protein family and was designated H-ray. Northern blot analysis shows that both genes are ubiquitously expressed. Using a bacterial expression vector, each clone was overexpressed in Escherichia coli. The two bacterially produced proteins possess GTP-binding activity.


Asunto(s)
Proteínas de Unión al GTP/genética , Músculo Esquelético/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , Escherichia coli/genética , Femenino , Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Técnicas In Vitro , Ratones , Datos de Secuencia Molecular , Embarazo , Ratas , Homología de Secuencia de Aminoácido , Distribución Tisular
19.
Mol Cell Biol ; 14(12): 8202-11, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7969157

RESUMEN

Mitogen-activated protein kinases, or extracellular signal-regulated kinases (ERKs), are serine/threonine protein kinases that are activated in response to a wide variety of extracellular stimuli and are encoded by a multigene family. Little is known about the function of the ERK-3 subfamily. To explore the molecular diversity of the ERK-3 subfamily, we isolated a novel human cDNA, designated Hu-ERK-3, from a fetal skeletal muscle library. Analysis of the complete 3,920-bp nucleotide sequence revealed that this clone encodes a predicted protein of 721 amino acids. In vitro transcription-translation generates a 97-kDa protein referred to as p97MAPK. Of all of the sequences compared, p97MAPK is the most homologous to rat ERK-3. Interestingly, although p97MAPK is highly (98%) homologous to ERK-3 at the amino acid level within the N-terminal two-thirds of the coding region, it diverges at the carboxyl terminus as a result of a unique extension of 178 amino acids. Although expression of p97MAPK was detected in all of the tissues tested by Northern (RNA) analysis, the most abundant expression was seen in skeletal muscle. An antibody raised against the unique C terminus recognized a 97-kDa protein in human cells. By using this antibody in an immune complex protein kinase assay, we have shown that treatment of human fibroblasts with serum or phorbol esters activates a myelin basic protein and histone H1 kinase activity in immunoprecipitates. p97MAPK appears to be the human homolog of rat ERK-3, and a member of this family is an active protein kinase.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Activadas por Mitógenos , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Clonación Molecular , Cartilla de ADN/química , Regulación de la Expresión Génica , Humanos , Técnicas In Vitro , Proteína Quinasa 6 Activada por Mitógenos , Datos de Secuencia Molecular , Músculos/enzimología , ARN Mensajero/genética , Mapeo Restrictivo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Distribución Tisular
20.
Am J Physiol ; 266(2 Pt 1): C351-9, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8141249

RESUMEN

Serine-threonine protein kinases in the ribosomal S6 kinase (rsk or p90rsk) family have been implicated as signaling intermediates in the cellular response to several growth factors. To investigate the molecular diversity of human p90rsk isoforms, mixed degenerate oligonucleotide polymerase chain reaction was used to isolate partial rsk cDNAs (1.1 kb). Three closely related human rsk cDNAs were obtained (HU-1, HU-2, HU-3). These cDNAs are encoded by separate genes based on DNA sequence diversity and distinct patterns seen with genomic Southern blots. Northern analysis revealed different sized mRNA transcripts for each isoform. A full-length HU-1 cDNA (3.1 kb) was subsequently isolated from a HeLa cell library. 5'-cDNA clones for HU-2 and HU-3 were isolated using the "rapid amplification of cDNA ends" strategy. Experiments using human x hamster somatic cell hybrids localized the HU-1 gene to human chromosome 3; HU-2 is on chromosome 6; and HU-3 is on the X chromosome. The tissue distribution of human rsk mRNAs was determined using ribonuclease protection assays. HU-3 mRNA was present in multiple RNA samples. HU-2 was expressed in fibroblast > muscle > lymphocyte = placenta > liver. HU-1 was expressed in Epstein-Barr virus lymphocyte > > muscle = liver > fat = placenta. These results indicate that the multiplicity of p90rsk isoforms is increased to at least three for humans and that marked tissue-/cell-specific differences in p90rsk isoform expression are present.


Asunto(s)
Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Northern Blotting , Southern Blotting , Clonación Molecular , Secuencia de Consenso , ADN Complementario/genética , Humanos , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Proteínas Quinasas S6 Ribosómicas , Distribución Tisular , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA