Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
Mais filtros

Medicinas Complementares
Intervalo de ano de publicação
1.
Crit Rev Oncog ; 26(2): 67-72, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34347973

RESUMO

K-Ras is one of the most important oncogenes in human oncogenesis. K-Ras transfection of normal rat fibroblasts induces phenotypic change from flat to round morphology. Then, we screened compounds inducing flat morphology in K-Ras transformed fibroblasts from microbial culture filtrates and plant extracts. As a result, the alkaloid conophylline was isolated from the leaves of Ervatamia microphylla collected in Thailand. Conophylline induced flat morphology and inhibited cellular invasion in K-Ras-transformed normal rat kidney (K-Ras-NRK) cells. It also inhibited the growth of the K-Ras-NRK tumor in mice. Cancer-associated fibroblasts are now considered to activate cancer growth. Conophylline was found to suppress secretions of various inflammatory cytokines by pancreatic cancer-associated fibroblasts. Moreover, when combined with gemcitabine, it inhibited the growth of pancreatic cancer growth in mice. Conophylline is orally active. Thus, the plant-derived alkaloid conophylline inhibited cancer growth directly and indirectly, and it shows promise as a new anticancer agent.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Alcaloides de Vinca , Animais , Genes ras , Camundongos , Folhas de Planta/química , Ratos , Tabernaemontana/química , Alcaloides de Vinca/farmacologia
2.
Lima; Instituto Nacional de Salud; ene. 2021.
Não convencional em Espanhol | BRISA | ID: biblio-1354015

RESUMO

INTRODUCCIÓN: Este documento técnico se realiza a solicitud del Hospital Santa Rosa, a través de la Gerencia de Riesgos y Evaluación de las Prestaciones del Seguro Integral de Salud; la pregunta PICO fue la siguiente: P: adultos con cáncer colorrectal metastásico de localización izquierda y genes RAS no mutados; I: cetuximab + quimioterapia (C+Qt); C: quimioterapia (Qt) o panitumumab + quimioterapia (P+Qt); O: Sobrevida global, sobrevida libre de progresión, tasa de respuesta y eventos adversos. a. Cuadro clínico: En Perú, el cáncer colorrectal constituye el quinto tipo de cáncer más frecuente y el sexto con mayor mortalidad. En el diagnóstico inicial, un 25% de pacientes presenta enfermedad metastásica distante. En los últimos veinte años, la sobrevida global de pacientes con cáncer colorrectal metastásico se ha extendido, gracias a la aprobación de nuevas terapias administradas en combinación con agentes citotóxicos y la identificación de mutaciones genéticas que pueden predecir la respuesta al tratamiento. b. Tecnología sanitaria: Cetuximab es un antagonista del receptor de factor del crecimiento epidérmico, indicado para el tratamiento del cáncer colorrectal metastásico en tumores con sobreexpresión del EGFR y de tipo KRAS no mutado. Actúa mediante inhibición del crecimiento celular, inducción de apoptosis y disminución de la producción de metaloproteinasas de matriz y del factor de crecimiento endotelial vascular. Se recomienda una dosis inicial de 400 mg/m2, seguida de dosis semanales de 250 mg/m2, administradas en monoterapia o en combinación con quimioterapia. Cuenta con aprobación de la FDA desde el año 2004. En Perú, cuenta con dos registros sanitarios con vigencia prorrogada provisional. OBJETIVO: Describir la evidencia científica disponible sobre la eficacia y seguridad de cetuximab para el tratamiento en primera línea de pacientes con cáncer colorrectal metastásico de localización izquierda y genes RAS no mutados. METODOLOGÍA: Búsqueda sistemática en Medline, EMBASE, The Cochrane Library y LILACS, complementada con la búsqueda en páginas de agencias gubernamentales y buscadores genéricos. La calidad se valoró usando AMSTAR 2 para revisiones sistemáticas (RS), la herramienta de la colaboración Cochrane para ensayos clínicos y AGREE II para las Guías de Práctica Clínica (GPC). RESULTADOS: Sobrevida global: C+Qt mejoró la sobrevida global, en comparación con solo Qt, tanto en combinación con FOLFIRI (hazard ratio [HR]: 0,65; IC 95%: 0,50 a 0,86; 28,7 meses vs. 21,7 meses), como con FOLFOX (HR: 0,69; IC 95%: 0,53 a 0,90; 22 meses vs. 18,7 meses). No se observó diferencias entre C+Qt y P+Qt. Sobrevida libre de progresión: C+Qt mejoró la sobrevida global, en comparación con solo Qt, tanto en combinación con FOLFIRI (HR: 0,50; IC 95%: 0,34 a 0,72; 12 meses vs. 8,9 meses) como con FOLFOX (HR: 0,68; IC 95%: 0,50 a 0,91; 9,2 meses vs. 7,6 meses). No se observó diferencias entre C+Qt y P+Qt. Tasa de respuesta objetiva: C+Qt produjo una tasa de respuesta objetiva significativamente más alta, en comparación con solo Qt, tanto en combinación con FOLFIRI (odds ratio [OR]: 3,99; IC 95%: 2,40 a 6,62), como con FOLFOX (OR: 2,60; IC 95%: 1,64 a 4,14). No se observó diferencias entre C+Qt y P+Qt. Eventos adversos grado: C+Qt incrementó hasta el doble la probabilidad de sufrir algún evento adverso, en comparación con solo Qt, con un riesgo incrementado de toxicidad dermatológica (RR: 20,76; IC 95%: 3,87 a 111,33), diarrea (RR: 1,48; IC 95%: 1,33 a 1,64), hipertensión (RR: 1,69; IC 95%: 1,17 a 2,46), anorexia (RR: 1,57; IC 95%: 1,18 a 2,10) y mucositis/estomatitis (RR: 2,69; IC 95%: 1,90 a 3,80). Recomendaciones en GPC: Sólo NCCN y ASCO incluyen recomendaciones específicas para cáncer colorrectal metastásico con tumores RAS no mutadas de localización izquierda. Ambas incluyen como opciones de tratamiento a C+Qt y P+Qt. Evaluaciones de Tecnología Sanitaria: Dos ETS nacionales no recomiendan dar cobertura a C+Qt en cáncer colorrectal metastásico RAS de tipo salvaje, citando aspectos relacionados con su eficacia y costo-efectividad. NICE recomienda, tanto C+Qt como P+Qt sin especificar la lateralidad del tumor. Evaluación de la calidad metodológica: Dos RS fueron consideradas como nivel de confianza bajo y dos como nivel de confianza críticamente bajo. Las GPC obtuvieron un puntaje en la valoración global de calidad que varío entre 60,1% y 84,6%. CONCLUSIONES: La evidencia sobre el uso de C+Qt para cáncer colorrectal de lado izquierdo es limitada. Los desenlaces de eficacia que comparan C+Qt provienen de análisis de subgrupos no especificados de dos ensayos clínicos, mientras que los desenlaces de seguridad provienen de evidencia que no considera la lateralidad del tumor. Las comparaciones entre C+Qt y P+Qt proceden de meta-análisis en red de comparaciones indirectas. En pacientes con cáncer colorrectal metastásico de lado izquierdo con genes RAS no mutados, la adición de cetuximab a la quimioterapia incrementó la sobrevida global alrededor de 7 meses cuando se combinó con FOLFIRI y alrededor de 3,3 meses cuando se combinó con FOLFOX. No se hallaron diferencias entre cetuximab y panitumumab. La sobrevida libre de progresión se incrementó en alrededor de 3,1 meses tras la adición de cetuximab a FOLFIRI y alrededor de 1,6 meses cuando se adicionó a FOLFOX. No se hallaron diferencias entre cetuximab y panitumumab. Los pacientes tratados con cetuximab más quimioterapia tuvieron el doble de probabilidad de sufrir algún evento adverso, en comparación con solo quimioterapia, siendo común observar un riesgo incrementado de presentar toxicidad dermatológica, diarrea, hipertensión, anorexia y mucositis/dermatitis. Cetuximab y panitumumab presentaron diferente perfil de eventos adversos. Sólo las GPC de NCCN y ASCO incluyen recomendaciones específicas para cáncer colorrectal metastásico con tumores RAS no mutados de localización izquierda. Ambas GPC incluyen como opciones de tratamiento a cetuximab y panitumumab acompañados por quimioterapia. Dos ETS nacionales recomiendan no dar cobertura a C+Qt en pacientes con cáncer colorrectal metastásico con tumores RAS de tipo salvaje, citando aspectos relacionados con su eficacia y costo-efectividad. La ETS de NICE recomienda, tanto C+Qt como P+Qt, para cáncer colorrectal metastásico con tumores RAS de tipo salvaje sin especificar la lateralidad del tumor. Dos RS fueron consideradas como nivel de confianza bajo y dos como nivel de confianza críticamente bajo. Las GPC obtuvieron un puntaje en la valoración global de calidad que varío entre 60,1% y 84,6%. Ambas RS fueron consideradas como nivel de confianza bajo. Las GPC incluidas obtuvieron un puntaje en la valoración global de calidad que varío grandemente entre 60,1% y 84,6%.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Genes ras , Cetuximab/uso terapêutico , Metástase Neoplásica/patologia , Eficácia , Análise Custo-Benefício
3.
Anticancer Res ; 40(7): 3819-3830, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32620621

RESUMO

BACKGROUND: Picrasma quassioides (PQ) is a traditional Asian herbal medicine with anti-tumor properties that can inhibit the viability of HepG2 liver cancer cells. H-Ras is often mutated in liver cancer, however, the effect of PQ treatment on H-Ras mutated liver cancer is unclear. This study aimed to investigate the role of PQ on ROS accumulation and mitochondrial dysfunction in H-ras mutated HepG2 (HepG2G12V) cells. MATERIALS AND METHODS: PQ ethanol extract-induced HepG2G12V apoptosis was analyzed by the MTT assay, fluorescence microscopy, flow cytometry and western blotting. RESULTS: PQ treatment affected cell migration and colony formation in HepG2G12V cells. Cleaved-caspase-3, cleaved-caspase-9 and BCL2 associated agonist of cell death (BAD) expression levels were increased, while the levels of B-cell lymphoma-extra large (Bcl-xL) were decreased with PQ treatment. PQ treatment led to a reduction of H-Ras expression levels in liver cancer cells, thus reducing their abnormal proliferation. Furthermore, it led to increased expression levels of Peroxiredoxin VI, which regulates the redox signal in cells. CONCLUSION: Taken together these results provide a new functional significance for the role of PQ in treating HepG2G12V liver cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Mitocôndrias Hepáticas/efeitos dos fármacos , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Espécies Reativas de Oxigênio/metabolismo , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Genes ras , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Picrasma/química , Proteínas Proto-Oncogênicas p21(ras)/biossíntese
4.
BMC Cancer ; 20(1): 687, 2020 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-32703200

RESUMO

BACKGROUND: First-line treatment with FOLFOXIRI plus bevacizumab (BEV) is highly effective and regarded as one of the standards-of-care for patients with metastatic colorectal cancer (mCRC), despite the high incidence of neutropenia and diarrhea as side effects. AXEPT, an Asian phase III study, showed that modified CAPIRI+BEV [capecitabine (CAP: 1600 mg/m2), irinotecan (IRI: 200 mg/m2), and BEV (7.5 mg/m2)] was non-inferior to FOLFIRI+BEV as a second-line therapy for mCRC patients and was associated with a lower incidence of hematologic toxicities. Thus, a reduced dose of the CAP and IRI regimen in combination with oxaliplatin (OX) and BEV (CAPOXIRI+BEV) may be more feasible than FOLFOXIRI+BEV, without compromising efficacy. METHODS: QUATTRO-II is an open-label, multicenter, randomized phase II study. In Step 1, the recommended doses of OX and IRI will be investigated as a safety lead-in. In Step 2, patients will be randomized to the recommended dose of either CAPOXIRI+BEV or FOLFOXIRI+BEV. Induction triplet chemotherapy plus BEV treatments will be administered for up to 4 months followed by fluoropyrimidine plus BEV maintenance. The primary endpoint is progression-free survival (PFS). The similarity in PFS between the two arms will be evaluated by observing whether the point estimate of hazard ratio (HR) for PFS falls between 0.80 and 1.25. Ensuring a 70% probability that the observed HR will be "0.8 < HR < 1.25" under the assumption of the true HR of 1.0, and 100 patients will be evaluated during the 3-year study period. Secondary endpoints include overall survival, overall response rate, safety, and patient reported outcome (PRO) (FACT/GOG-Ntx4). DISCUSSION: Considering the lower incidence of hematologic toxicities with modified CAPIRI+BEV than with FOLFIRI+BEV, CAPOXIRI+BEV may be a promising treatment option if sufficient efficacy and lower hematologic toxicities are indicated in this study. Additionally, a lower incidence of peripheral sensory neuropathy (PSN) reported following CAPEOX treatment compared to that after FOLFOX in ACHIEVE, an adjuvant phase III trial, suggest that CAPOXIRI+BEV can mitigate OX-induced PSN. TRIAL REGISTRATION: Clinicaltrials.gov NCT04097444 . Registered September 20, 2019, https://clinicaltrials.gov/ct2/show/study/NCT04097444 / Japan Registry of Clinical Trials jRCTs041190072. Registered October 9, 2019.


Assuntos
Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bevacizumab/administração & dosagem , Camptotecina/análogos & derivados , Neoplasias do Colo/tratamento farmacológico , Desoxicitidina/análogos & derivados , Fluoruracila/análogos & derivados , Neoplasias Retais/tratamento farmacológico , Adenocarcinoma/patologia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Camptotecina/administração & dosagem , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Desoxicitidina/administração & dosagem , Esquema de Medicação , Fluoruracila/administração & dosagem , Genes ras , Glucuronosiltransferase/genética , Humanos , Leucovorina/administração & dosagem , Compostos Organoplatínicos/administração & dosagem , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Retais/genética , Neoplasias Retais/patologia
5.
Int J Toxicol ; 39(3): 198-206, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32372678

RESUMO

The Tg.rasH2 mouse was developed as an alternative model to the traditional 2-year mouse bioassay for pharmaceutical carcinogenicity testing. This model has found extensive use in support of pharmaceutical drug development over the last few decades. It has the potential to improve quality and timeliness, reduce animal usage, and in some instances allow expedient decision-making regarding the human carcinogenicity potential of a drug candidate. Despite the increased use of the Tg.rasH2 model, there has been no systematic survey of current practices in the design, interpretation of results from the bioassay, and global health authority perspectives. Therefore, the aim of this work was to poll the pharmaceutical industry on study design practices used in the dose range finding and definitive 6-month studies and on results relative to the ongoing negotiations to revise The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use S1 Guidance. Twenty-two member companies of International Consortium for Innovation and Quality in Pharmaceutical Development DruSafe Leadership Group participated in the survey, sharing experiences from studies conducted with 55 test compounds between 2010 and 2018. The survey results provide very useful insights into study design and interpretation. Importantly, the results identified several key opportunities for reducing animal use and increasing the value of testing for potential human carcinogenicity using this model. Recommended changes to study designs that would reduce animal usage include eliminating the requirement to include positive control groups in every study, use of nontransgenic wild-type littermates in the dose range finding study, and use of microsampling to reduce or eliminate satellite groups for toxicokinetics.


Assuntos
Testes de Carcinogenicidade/métodos , Carcinógenos/toxicidade , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Indústria Farmacêutica/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Animais , Bioensaio , Genes ras , Camundongos Transgênicos , Projetos de Pesquisa , Inquéritos e Questionários
6.
Cancer Cell ; 37(1): 3-4, 2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31951561

RESUMO

Drugs that target KRAS 12C covalently, AMG 510 and MRTX849, are now in the clinic. Recent papers describe development of these compounds, their selectivity and properties, early clinical data, and potential combination therapies. These papers herald a new era in Ras research, with improved drugs and strategies certain to follow.


Assuntos
Antineoplásicos/farmacologia , Genes ras , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Piridinas/farmacologia , Pirimidinas/farmacologia , Animais , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Camundongos , Mutação , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Pesquisa Translacional Biomédica , Resultado do Tratamento
7.
Nutr Cancer ; 70(7): 1075-1087, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30273070

RESUMO

Pancreatic cancer (PC) patients have poor prognosis and survival rate. Gemcitabine, the drug of choice has a dismal 15% response rate. Earlier, we reported that Garcinol alone and in combination with gemcitabine showed a dose-dependent favorable response on PC cell lines. This study probes the in vivo effects of dietary Garcinol on PC progression in transgenic PC mice (KPC; K-ras and p53 conditional mutant). KPC male mice were divided into: KC- Control diet; KGr-0.05% Garcinol diet; KGm-Gemcitabine injected; KGG - Garcinol diet + Gemcitabine injected groups. Changes in tumor progression, toxicity, or cell morphology were monitored by magnetic resonance imaging, Fore-stomach, and blood smear, respectively. Pancreatic Intraepithelial Neoplasia (mPanIN) grading with hematoxylin and eosin (H&E) staining was conducted on pancreas and validated by immunohistochemistry. The KGr group showed improved survival, no observable toxicity with marked reduction in papilloma formation in the fore-stomach, and a higher ratio of NK and NKT cells compared to Non-NK lymphocytes. Additionally, the KGr, KGm, and KGG groups showed reduction in tumor volumes and reduced number of advanced mouse PanIN3. Dietary Garcinol alone and in combination with gemcitabine retarded the progression of PC in transgenic PC mice, arresting the cancer in the earlier stages, improving prognosis and survival.


Assuntos
Neoplasias Pancreáticas/dietoterapia , Terpenos/farmacologia , Animais , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Suplementos Nutricionais , Genes p53 , Genes ras , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/tratamento farmacológico , Subunidade beta da Proteína Ligante de Cálcio S100/imunologia , Proteína Smad4/imunologia , Taxa de Sobrevida , Terpenos/efeitos adversos , Gencitabina
8.
J Vasc Interv Radiol ; 29(8): 1194-1202.e1, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29887183

RESUMO

PURPOSE: This study used the Oncopig Cancer Model (OCM) to develop alcohol-induced fibrosis in a porcine model capable of developing hepatocellular carcinoma. MATERIALS AND METHODS: Liver injury was induced in 8-week-old Oncopigs (n = 10) via hepatic transarterial infusion of 0.75 mL/kg ethanol-ethiodized oil (1:3 v/v). Feasibility was assessed in an initial Oncopig cohort (n = 5) by histologic analysis at 8 weeks after induction, and METAVIR results were compared to age- and sex-matched healthy controls (n = 5). Liver injury was then induced in a second OCM cohort (n = 5) for a time-course study, with post-induction disease surveillance via biweekly physical exam, lab analysis, and liver biopsies until 20 weeks after induction. RESULTS: In Cohort 1, 8-week post-induction liver histologic analysis revealed median METAVIR F3 (range, F3-F4) fibrosis, A2 (range, A2-A3) inflammation, and 15.3% (range, 5.0%-22.9%) fibrosis. METAVIR and inflammation scores were generally elevated compared to healthy controls (F0-F1, P = 0.0013; A0-A1, P = .0013; median percent fibrosis 8.7%, range, 5.8%-12.1%, P = .064). In Cohort 2, histologic analysis revealed peak fibrosis severity of median METAVIR F3 (range, F2-F3). However, lack of persistent alcohol exposure resulted in liver recovery, with median METAVIR F2 (range, F1-F2) fibrosis at 20 weeks after induction. No behavioral or biochemical abnormalities were observed to indicate liver decompensation. CONCLUSIONS: This study successfully validated a protocol to develop METAVIR F3-F4 fibrosis within 8 weeks in the OCM, supporting its potential to serve as a model for hepatocellular carcinoma in a fibrotic liver background. Further investigation is required to determine if repeated alcohol liver injury is required to develop an irreversible METAVIR grade F4 porcine cirrhosis model.


Assuntos
Carcinoma Hepatocelular/etiologia , Transformação Celular Neoplásica/patologia , Etanol , Óleo Etiodado , Cirrose Hepática Alcoólica/etiologia , Neoplasias Hepáticas/etiologia , Fígado/patologia , Animais , Animais Geneticamente Modificados , Biópsia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Progressão da Doença , Feminino , Genes p53 , Genes ras , Cirrose Hepática Alcoólica/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Índice de Gravidade de Doença , Sus scrofa , Fatores de Tempo
9.
Sci Rep ; 8(1): 6409, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29686295

RESUMO

Signal transducer and activator of transcription 3 (STAT3) is a transcription factor that is latent but constitutively activated in many types of cancers. It is well known that STAT3 plays a key role in inflammation-associated tumorigenesis. Curcumin is an anti-inflammatory natural compound isolated from the turmeric (Curcuma longa L., Zingiberaceae) that has been extensively used in a traditional medicine over the centuries. In the present study, we have found that curcumin inhibits STAT3 signaling that is persistently overactivated in H-Ras transformed breast epithelial cells (H-Ras MCF10A). Specific cysteine residues present in STAT3 appear to be critical for the activity as well as conformation of this transcription factor. We identified the cysteine residue 259 of STAT3 as a putative site for curcumin binding. Site-directed mutation of this cysteine residue abolished curcumin-induced inactivation of STAT3 and apoptosis in H-Ras MCF10A cells. The α,ß-unsaturated carbonyl moiety of curcumin appears to be essential in its binding to STAT3 in H-Ras MCF10A cells. Tetrahydrocurcumin that lacks such electrophilic moiety failed to interact with STAT3 and to induce apoptosis in the same cell line. Taken together, our findings suggest that curcumin can abrogate aberrant activation of STAT3 through direct interaction, thereby inhibiting STAT3-mediated mammary carcinogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Curcumina/metabolismo , Curcumina/farmacologia , Cisteína/metabolismo , Genes ras , Glândulas Mamárias Humanas/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Linhagem Celular Transformada , Curcumina/análogos & derivados , DNA/metabolismo , Dimerização , Humanos , Glândulas Mamárias Humanas/patologia , Fator de Transcrição STAT3/química , Compostos de Sulfidrila/metabolismo , Transcrição Gênica
10.
Eur J Cancer ; 92: 40-47, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29413688

RESUMO

BACKGROUND: The prognosis of poorly differentiated thyroid carcinomas (PDTC) is heterogeneous though generally poor. The objectives of this study were to identify clinical and molecular factors of poor prognosis. METHODS: One hundred four consecutive patients treated for a PDTC between 01/01/2000 and 31/12/2010 were included in this study. A pathological review was done for all cases (blinded to clinical data and outcome). RESULTS: All patients underwent thyroidectomy. Adjuvant radioactive-iodine was administered in 95.2% of them. Tumours were pT3 or pT4 in 68.3% of cases and metastatic in 38.5% of patients. Extrathyroidal extension (ETE) was observed in 40% of patients. At the end of the initial treatment, only 37% of patients were considered in remission. Fifty-two patients (50%) became refractory to radioiodine during follow-up. The 5-year overall survival was 72.8% and the 5-year recurrence-free survival (RFS) was 45.3%. Remission after initial treatment was an independent factor of RFS (HR = 0.22; [0.10-0.49]). ETE was the only significant parameter influencing the overall survival in multivariate analysis. TERT promoter mutations at positions -124 (C228T) and -146 (C250T) were present in 38.1% of analysed patients and significantly associated with radioiodine resistance but not with overall survival. Half of TERT promoter mutant tumours harboured also RAS or BRAF mutations. CONCLUSION: PDTC form a heterogeneous group of patients with usual late-stage diagnosis, low radioactive iodine avidity and frequent metastatic spread. TERT promoter mutations could help to identify patients with high risk of radio-iodine refractoriness.


Assuntos
Carcinoma/secundário , Carcinoma/cirurgia , Diferenciação Celular , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/cirurgia , Tireoidectomia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Carcinoma/genética , Carcinoma/mortalidade , Criança , Progressão da Doença , Intervalo Livre de Doença , Feminino , França , Genes ras , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Mutação , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Proteínas Proto-Oncogênicas B-raf/genética , Tolerância a Radiação , Radioterapia Adjuvante , Fatores de Risco , Telomerase/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/mortalidade , Tireoidectomia/efeitos adversos , Tireoidectomia/mortalidade , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
11.
Oncotarget ; 8(27): 44295-44311, 2017 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-28574828

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease in urgent need of newer therapeutic modalities. Majority of patients with PDAC have mutations in KRAS, which unfortunately remains an ineffectual target. Our strategy here is to target KRAS downstream effectors PI3K and mTOR. In this study, we investigated the antitumor efficacy of the novel PI3K and mTOR dual inhibitor VS-5584 in PDAC. Our data shows that PI3K/mTOR dual inhibition causes ERK activation in all tested PDAC cell lines. Although the MEK inhibitor GSK1120212 could abrogate VS-5584-induced ERK activation, it did not substantially enhance cell death in all the cell lines tested. However, combination with ERK inhibitor SCH772984 not only mitigated VS-5584-induced ERK activation but also enhanced VS-5584-induced cell death. In a xenograft model of PDAC, we observed 28% and 44% tumor inhibition for individual treatment with VS-5584 and SCH772984, respectively, while the combined treatment showed superior tumor inhibition (80%) compared to vehicle control treatment. Our findings support the clinical development of VS-5584 and ERK inhibitor combination for PDAC treatment.


Assuntos
Antineoplásicos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Morfolinas/farmacologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genes ras , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Rev Neurol ; 64(s03): S13-S17, 2017 May 17.
Artigo em Espanhol | MEDLINE | ID: mdl-28524213

RESUMO

INTRODUCTION: The term 'RASopathies' covers a series of diseases that present mutations in the genes that code for the proteins of the RAS/MAPK pathway. These diseases include neurofibromatosis type 1, Noonan syndrome, Legius syndrome, LEOPARD syndrome, Costello syndrome and cardiofaciocutaneous syndrome. Involvement of the RAS/MAPK pathway not only increases predisposition to develop tumours, but also determines the presence of phenotypic anomalies and alterations in learning processes. AIM: To review the use of therapeutic strategies with mechanisms that have a selective action on RASopathies. DEVELOPMENT: The fact that the RAS pathway is involved in a third of all neoplasms has led to the development and study of different drugs at this level. Some of these pharmaceutical agents have been tested in RASopathies, mainly in neurofibromatosis type 1. Here we analyse the use of different antitarget treatments: drugs that act on the membrane receptors, such as tyrosine kinase inhibitors, in the mTOR pathway or MEK inhibitors. These latter have shown potential benefits in recent studies conducted on different RASopathies. CONCLUSIONS: Today, thanks to the results from the first studies conducted with MEK inhibitor based mainly on animal models, a number of promising clinical trials are being carried out.


TITLE: Actualizacion del tratamiento de las rasopatias.Introduccion. El termino 'rasopatias' agrupa una serie de enfermedades que presentan mutaciones en genes que codifican las proteinas de la via RAS/MAPK. Estas enfermedades incluyen la neurofibromatosis de tipo 1, el sindrome de Noonan, el sindrome de Legius, el sindrome LEOPARD, el sindrome de Costello y el sindrome cardiofaciocutaneo. La afectacion de la via RAS/MAPK no solo aumenta la predisposicion a desarrollar tumores, sino que tambien determina la presencia de anomalias fenotipicas y alteraciones en los procesos de aprendizaje. Objetivo. Revisar el papel del uso de estrategias terapeuticas con mecanismos de accion selectivo en las rasopatias. Desarrollo. El hecho de que la via RAS participe en un tercio de las neoplasias ha motivado el desarrollo y el estudio de distintos farmacos a este nivel. Algunos de estos farmacos han sido probados en las rasopatias, principalmente en la neurofibromatosis de tipo 1. Analizamos el uso de distintos tratamientos antidiana: farmacos que actuan en los receptores de membrana, como los inhibidores de la tirosincinasa, en la via mTOR o los inhibidores de MEK. Existe un potencial beneficio de estos ultimos en estudios recientes realizados en distintas rasopatias. Conclusiones. Actualmente, gracias a los resultados de los primeros trabajos desarrollados con inhibidor de MEK basados principalmente en modelos animales, se estan realizando multiples ensayos clinicos prometedores.


Assuntos
Anormalidades Múltiplas/tratamento farmacológico , Genes ras , Doenças Genéticas Inatas/tratamento farmacológico , Sistema de Sinalização das MAP Quinases , Terapia de Alvo Molecular , Proteínas ras/antagonistas & inibidores , Anormalidades Múltiplas/classificação , Anormalidades Múltiplas/genética , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Doenças Genéticas Inatas/classificação , Doenças Genéticas Inatas/genética , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Síndromes Neoplásicas Hereditárias/tratamento farmacológico , Síndromes Neoplásicas Hereditárias/genética , Neurofibromatose 1/tratamento farmacológico , Neurofibromatose 1/genética , Síndrome de Noonan/tratamento farmacológico , Síndrome de Noonan/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Síndrome , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas ras/genética
13.
Am J Chin Med ; 45(3): 423-441, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28359198

RESUMO

It has been shown that plumbagin, a bioactive naphthoquinone isolated from three major plant families viz. Plumbaginaceae, Ebenceae and Droseraceae, definitively exhibits anticancer potential in diverse cancer cells both in vitro and in vivo. Plumbagin shows antineoplastic effects via multi-channel molecular mechanisms, including the induction of apoptosis and autophagy, the disruption of the cell cycle, the inhibition of invasion and metastasis, and anti-angiogenesis. Plumbagin inhibits the growth of cancer cells mainly through the modulation of the signals of PI3K/Akt/mTOR, AMPK, Ras, and so on. The pharmaceutical applications of plumbagin combined with nanocarriers to achieve better therapeutic efficiency are discussed in this review Among them, liposomes, nanoparticles, microspheres, micelles, and nisosomes are used in cancer treatment. The anticancer study of plumbagin in vivo is also summarized in this review. On the whole, we aim to review the research progress of plumbagin both in pharmacological and pharmaceutical filed, which may provide some reference for further research of plumbagin.


Assuntos
Antineoplásicos Fitogênicos , Naftoquinonas/farmacologia , Naftoquinonas/uso terapêutico , Neoplasias/tratamento farmacológico , Fitoterapia , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Droseraceae/química , Ebenaceae/química , Genes ras , Humanos , Técnicas In Vitro , Camundongos , Naftoquinonas/isolamento & purificação , Neoplasias/genética , Neoplasias/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Plumbaginaceae/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo
14.
Sci Rep ; 7: 40153, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28065941

RESUMO

Di(2-ethylhexyl) phthalate (DEHP), as a widespread environmental pollutant and an endocrine disruptor, can disturb the homeostasis of thyroid hormones (THs). In order to elucidate roles of the MAPK and PI3K/Akt pathways and hepatic enzymes in thyroid-disrupting effects of DEHP, Sprague-Dawley rats were dosed with DEHP by gavage for 30 consecutive days; Nthy-ori 3-1 cells were treated with DEHP with NAC, k-Ras siRNA or inhibitors (U0126 and wortmannin). Results showed that DEHP led to histopathologic changes in rat thyroid and liver, such as the decrease in thyroid follicular cavity diameter, hepatocyte edema. Triiodothyronine (T3), thyroxine (T4) and thyrotropin releasing hormone (TRH) were reduced. DEHP caused ROS production, oxidative stress and k-Ras upregulation, thereby activating the ERK and Akt pathways in vivo and in vitro. Moreover, TRH receptor (TRHr) level was elevated after the activation of the Akt pathway and was downregulated after the inhibition of the Akt pathway. However, TRHr was not modulated by the ERK pathway. Additionally, hepatic enzymes, including Ugt1a1, CYP2b1, Sult1e1, and Sult2b1, were significantly induced after DEHP exposure. Taken together, DEHP can perturb TH homeostasis and reduce TH levels. The activated Ras/Akt/TRHr pathway and induced hepatic enzymes play vital roles in thyroid-disrupting effects of DEHP.


Assuntos
Dietilexilftalato/toxicidade , Disruptores Endócrinos/toxicidade , Homeostase/efeitos dos fármacos , Fígado/efeitos dos fármacos , Transdução de Sinais , Hormônios Tireóideos/metabolismo , Animais , Células Cultivadas , Genes ras , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Fígado/enzimologia , Fígado/patologia , Sistema de Sinalização das MAP Quinases , Masculino , Estresse Oxidativo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Células Epiteliais da Tireoide/efeitos dos fármacos , Glândula Tireoide/patologia , Glândula Tireoide/ultraestrutura
15.
PLoS One ; 11(7): e0159531, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27427904

RESUMO

Transcription factors have an important role in cancer but are difficult targets for the development of tumour therapies. These factors include the Ets family, and in this study Elk3 that is activated by Ras oncogene /Erk signalling, and is involved in angiogenesis, malignant progression and epithelial-mesenchymal type processes. We previously described the identification and in-vitro characterisation of an inhibitor of Ras / Erk activation of Elk3 that also affects microtubules, XRP44X. We now report an initial characterisation of the effects of XRP44X in-vivo on tumour growth and metastasis in three preclinical models mouse models, subcutaneous xenografts, intra-cardiac injection-bone metastasis and the TRAMP transgenic mouse model of prostate cancer progression. XRP44X inhibits tumour growth and metastasis, with limited toxicity. Tumours from XRP44X-treated animals have decreased expression of genes containing Elk3-like binding motifs in their promoters, Elk3 protein and phosphorylated Elk3, suggesting that perhaps XRP44X acts in part by inhibiting the activity of Elk3. Further studies are now warranted to develop XRP44X for tumour therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , Piperazinas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Proteínas Proto-Oncogênicas c-ets/antagonistas & inibidores , Pirazóis/farmacologia , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Feminino , Genes ras/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Microtúbulos/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Ratos , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Planta Med ; 82(11-12): 1096-104, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27280936

RESUMO

Three new rotenoids (1-3), two new isoflavonoids (4 and 5), and six known analogues (6-11) were isolated from an n-hexane partition of a methanol extract of the fruits of Millettia caerulea, with the structures of the new compounds elucidated by analysis of their spectroscopic data. The relative configurations of the rotenoids were determined by interpretation of their NMR spectroscopic data, and their absolute configurations were established using electronic circular dichroism spectra and specific rotation values. All compounds isolated were evaluated for their cell growth inhibitory activity against the HT-29 human colon cancer cell line, and the known compounds, (-)-3-hydroxyrotenone (6) and (-)-rotenone (7), were found to be potently active. When tested in an NF-κB inhibition assay, compound 6 showed activity. This compound, along with the new compound, (-)-caeruleanone D (1), and the known compound, ichthynone (8), exhibited K-Ras inhibitory potency. Further bioactivity studies showed that the new compounds, (-)-3-deoxycaeruleanone D (2) and (-)-3-hydroxycaeruleanone A (3), and the known compounds 8 and 11 induced quinone reductase in murine Hepa 1c1c7 cells.


Assuntos
Isoflavonas/isolamento & purificação , Millettia/química , Animais , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Indução Enzimática/efeitos dos fármacos , Frutas/química , Genes ras/efeitos dos fármacos , Células HT29 , Células HeLa , Humanos , Isoflavonas/química , Isoflavonas/farmacologia , Camundongos , Estrutura Molecular , NAD(P)H Desidrogenase (Quinona)/metabolismo , NF-kappa B/antagonistas & inibidores , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Rotenona/química
17.
Clin. transl. oncol. (Print) ; 18(6): 608-616, jun. 2016. tab, ilus, graf
Artigo em Inglês | IBECS | ID: ibc-152756

RESUMO

Background: Although Ras-association domain family of gene 2 (RASSF2) has been shown to undergo promoter methylation at high frequency in some cancer types and in brain metastases, its clinical utility as a useful prognostic molecular marker remains unclear in gastric cancer. Methods: Prognostic significance of RASSF2 expression was retrospectively analysed by immunohistochemically in 105 patients with gastric cancer who underwent curative gastrectomy. Results: Low RASSF2 expression was detected in 58 (55 %) patients, whereas 47 patients (45 %) had high RASSF2 expression. Lymph node involvement, pT stage, TNM stage, vascular invasion, perineural invasion and the presence of recurrence were found to be significantly related to RASSF2 expression levels. Low PRL-3 expression was closely correlated with lymph node metastasis (p = 0.001), advanced pT stage (p = 0.021), advanced TNM stage (p < 0.001), the presence of vascular invasion (p < 0.001), perineural invasion (p = 0.018) and high prevalence of recurrence (p = 0.003) compared with high RASSF2 expression. The median disease-free survival (DFS) time for patients with low RASSF2 expression was significantly worse than that of patients with high RASSF2 expression (10.2 vs. 50.6 months, p < 0.001). In addition, patients with high RASSF2 expression had the higher overall survival (OS) interval compared to patients with low RASSF2 expression (NR vs. 14.9 months, p < 0.001). In the multivariate analysis, the rate of RASSF2 expression levels was an independent prognostic factor, for DFS [p < 0.001, HR 0.12 (0.10-0.88)] and OS [p < 0.001, HR 0.10 (0.04-0.46)], as were pT stage and TNM stage, respectively. Conclusions: RASSF2 may be an important molecular marker for carcinogenesis, prognosis and progression in gastric cancer, but the potential value of RASSF2 expression as a useful molecular marker in gastric cancer progression should be evaluated, comprehensively. It would be possible to develop treatments targeting RASSF2 and advance new treatment strategies for gastric cancer


No disponible


Assuntos
Humanos , Masculino , Feminino , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Prognóstico , Gastrectomia/métodos , Genes ras , Proteínas ras/análise , Quimioterapia Adjuvante , Estudos Retrospectivos , Carcinoma/diagnóstico , Carcinoma/tratamento farmacológico , Quimiorradioterapia/métodos , Quimiorradioterapia , Leucovorina/uso terapêutico , Fluoruracila/uso terapêutico
18.
Proc Natl Acad Sci U S A ; 113(11): 3078-83, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929329

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterized by an exuberant inflammatory desmoplastic response. The PDAC microenvironment is complex, containing both pro- and antitumorigenic elements, and remains to be fully characterized. Here, we show that sensory neurons, an under-studied cohort of the pancreas tumor stroma, play a significant role in the initiation and progression of the early stages of PDAC. Using a well-established autochthonous model of PDAC (PKC), we show that inflammation and neuronal damage in the peripheral and central nervous system (CNS) occurs as early as the pancreatic intraepithelial neoplasia (PanIN) 2 stage. Also at the PanIN2 stage, pancreas acinar-derived cells frequently invade along sensory neurons into the spinal cord and migrate caudally to the lower thoracic and upper lumbar regions. Sensory neuron ablation by neonatal capsaicin injection prevented perineural invasion (PNI), astrocyte activation, and neuronal damage, suggesting that sensory neurons convey inflammatory signals from Kras-induced pancreatic neoplasia to the CNS. Neuron ablation in PKC mice also significantly delayed PanIN formation and ultimately prolonged survival compared with vehicle-treated controls (median survival, 7.8 vs. 4.5 mo; P = 0.001). These data establish a reciprocal signaling loop between the pancreas and nervous system, including the CNS, that supports inflammation associated with oncogenic Kras-induced neoplasia. Thus, pancreatic sensory neurons comprise an important stromal cell population that supports the initiation and progression of PDAC and may represent a potential target for prevention in high-risk populations.


Assuntos
Capsaicina/uso terapêutico , Carcinoma Ductal Pancreático/prevenção & controle , Denervação , Pâncreas/inervação , Neoplasias Pancreáticas/prevenção & controle , Células Receptoras Sensoriais/fisiologia , Adenocarcinoma in Situ/patologia , Adenocarcinoma in Situ/fisiopatologia , Vias Aferentes , Animais , Animais Recém-Nascidos , Capsaicina/administração & dosagem , Capsaicina/farmacologia , Carcinoma Ductal Pancreático/etiologia , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/fisiopatologia , Carcinoma Ductal Pancreático/terapia , Ceruletídeo/toxicidade , Progressão da Doença , Feminino , Gânglios Simpáticos/fisiopatologia , Genes ras , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mielite/complicações , Mielite/genética , Mielite/fisiopatologia , Invasividade Neoplásica , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/fisiopatologia , Neoplasias Pancreáticas/terapia , Pancreatite/induzido quimicamente , Pancreatite/complicações , Pancreatite/fisiopatologia , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/complicações , Lesões Pré-Cancerosas/fisiopatologia , Células Receptoras Sensoriais/efeitos dos fármacos , Medula Espinal/fisiopatologia , Tratos Espinotalâmicos/fisiopatologia , Vértebras Torácicas
19.
Tumour Biol ; 37(7): 8811-24, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26747179

RESUMO

The CD44 isoform containing variant exon v6 (CD44v6) plays an important role in the progression, metastasis, and prognosis of colorectal cancer (CRC). Recently, it was found that CD44v6 is involved in acquired drug resistance. This study aimed to investigate the molecular mechanism of CD44v6 in the resistance of CRC cells to chemotherapy. A stable CD44v6 overexpression model in SW480 cells was established via lentiviral transduction. The chemosensitivity of cells to 5-fluorouracil (5-FU) and oxaliplatin (L-OHP) was determined by cell counting kit (CCK)-8, lactate dehydrogenase (LDH) release, and colony formation assays. Immunohistochemical staining of CD44v6 was performed in human CRC tissues. The key components in cell apoptosis, drug efflux and metabolism, mismatch repair, autophagy, epithelial-mesenchymal transition (EMT), and the PI3K-Akt and MAPK-Ras-Erk1/2 pathways were assessed using flow cytometry, quantitative real-time polymerase chain reaction (PCR), and western blot assays. The CD44v6 overexpression cells showed a higher viability, a lower LDH release rate, and an increased clonogenicity than the control cells under drug treatment. Moreover, overexpression of CD44v6 resulted in enhanced autophagy flux, EMT, and phosphorylation of Akt and Erk in the presence of drugs. Furthermore, high CD44v6 expression in the primary tumor was closely associated with an early recurrence in CRC patients who underwent curative surgery and adjuvant chemotherapy. In conclusion, overexpression of CD44v6 contributes to chemoresistance in SW480 cells under cytotoxic stress via the modulation of autophagy, EMT, and activation of the PI3K-Akt and MAPK-Ras-Erk pathways.


Assuntos
Autofagia/genética , Neoplasias do Colo/genética , Resistencia a Medicamentos Antineoplásicos/genética , Receptores de Hialuronatos/genética , Regulação para Cima/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Fluoruracila/farmacologia , Genes ras/genética , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/genética , Estudos Retrospectivos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Regulação para Cima/efeitos dos fármacos
20.
Clin Cancer Res ; 22(2): 374-82, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26307133

RESUMO

PURPOSE: Copy number alterations have been shown to be involved in melanoma pathogenesis. The randomized phase III clinical trial E2603: carboplatin, paclitaxel, ± sorafenib (CP vs. CPS) offers a large collection of tumor samples to evaluate association of somatic mutations, genomic alterations, and clinical outcomes, prior to current FDA-approved therapies. EXPERIMENTAL DESIGN: Copy number and mutational analysis on 119 pretreatment samples was performed. RESULTS: CPS therapy was associated with improved progression-free survival (PFS) compared with CP in patients with tumors with RAF1 (cRAF) gene copy gains (HR, 0.372; P = 0.025) or CCND1 gene copy gains (HR, 0.45; P = 0.035). CPS therapy was associated with improved overall survival (OS) compared with CP in patients with tumors with KRAS gene copy gains (HR, 0.25; P = 0.035). BRAF gene copy gain and MET amplification were more common in samples with V600K versus V600E mutations (P < 0.001), which was validated in The Cancer Genome Atlas (TCGA) dataset. CONCLUSIONS: We observed improved treatment response with CPS in patients with melanoma whose tumors have RAF1 (cRAF), KRAS, or CCND1 amplification, all of which can be attributed to sorafenib targeting CRAF. These genomic alterations should be incorporated in future studies for evaluation as biomarkers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carboplatina/uso terapêutico , Variações do Número de Cópias de DNA/efeitos dos fármacos , Melanoma/tratamento farmacológico , Melanoma/genética , Niacinamida/análogos & derivados , Paclitaxel/uso terapêutico , Compostos de Fenilureia/uso terapêutico , Variações do Número de Cópias de DNA/genética , Análise Mutacional de DNA/métodos , Intervalo Livre de Doença , Método Duplo-Cego , Genes ras/genética , Humanos , Mutação/efeitos dos fármacos , Mutação/genética , Estadiamento de Neoplasias/métodos , Niacinamida/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas c-met/genética , Sorafenibe , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA