Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Intervalo de ano de publicação
1.
J Nat Med ; 78(2): 342-354, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38324123

RESUMO

Evodiamine, a novel alkaloid, was isolated from the fruit of tetradium. It exerts a diversity of pharmacological effects and has been used to treat gastropathy, hypertension, and eczema. Several studies reported that evodiamine has various biological effects, including anti-nociceptive, anti-bacterial, anti-obesity, and anti-cancer activities. However, there is no research regarding its effects on drug-resistant cancer. This study aimed to investigate the effect of evodiamine on human vemurafenib-resistant melanoma cells (A375/R cells) proliferation ability and its mechanism. Cell activity was assessed using the cell counting kit-8 (CCK-8) method. Flow cytometry assay was used to assess cell apoptosis and cell cycle. A xenograft model was used to analyze the inhibitory effects of evodiamine on tumor growth. Bioinformatics analyses, network pharmacology, and molecular docking were used to explore the potential mechanism of evodiamine in vemurafenib-resistant melanoma. RT-qPCR and Western blotting were performed to reveal the molecular mechanism. The alkaloid extract of the fruit of tetradium, evodiamine showed the strongest tumor inhibitory effect on vemurafenib-resistant melanoma cells compared to treatment with vemurafenib alone. Evodiamine inhibited vemurafenib-resistant melanoma cell growth, proliferation, and induced apoptosis, conforming to a dose-effect relationship and time-effect relationship. Results from network pharmacology and molecular docking suggested that evodiamine might interact with IRS4 to suppress growth of human vemurafenib-resistant melanoma cells. Interestingly, evodiamine suppressed IRS4 expression and then inhibited PI3K/AKT signaling pathway, and thus had the therapeutic action on vemurafenib-resistant melanoma.


Assuntos
Alcaloides , Antineoplásicos , Melanoma , Quinazolinas , Humanos , Vemurafenib/farmacologia , Vemurafenib/uso terapêutico , Melanoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Simulação de Acoplamento Molecular , Transdução de Sinais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Proliferação de Células , Alcaloides/farmacologia , Linhagem Celular Tumoral , Proteínas Substratos do Receptor de Insulina/metabolismo
2.
Biol Direct ; 19(1): 6, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38178263

RESUMO

BACKGROUND: The outcome of Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) remain dismal despite the development of treatment. Targeted therapy is gaining more and more attention in improving prognosis. METHODS: Expression of BRAF was analyzed by RT-qPCR in AML and MDS patients. Cells viability treated by drugs was measured by CCK-8 assay. Network pharmacology and RNA-sequence were used to analyze the mechanism of drugs and verified in vitro and xenograft tumor model. RESULTS: Here we showed that BRAF was overexpressed in AML and MDS patients, and correlated with poor prognosis. The BRAF inhibitor-Vemurafenib (VEM) could significantly induce senescence, proliferation inhibition and apoptosis in AML cells, which can be enhanced by Bortezomib (BOR). This inhibitory effect was also verified in CD34 + cells derived from AML patients. Mechanistically, we showed that VEM combined with BOR could turn on HIPPO signaling pathway, thereby inducing cellular senescence in AML cells and xenograft mouse. CONCLUSIONS: Taken together, our findings demonstrate a significant upregulation of BRAF expression in AML and MDS patients, which is associated with unfavorable clinical outcomes. We also discovered that the BRAF inhibitor Vemurafenib induces cellular senescence through activation of the HIPPO signaling pathway. Analysis of BRAF expression holds promise as a prognostic indicator and potential therapeutic target for individuals with AML and MDS.


Assuntos
Leucemia Mieloide Aguda , Síndromes Mielodisplásicas , Humanos , Animais , Camundongos , Vemurafenib/farmacologia , Vemurafenib/uso terapêutico , Via de Sinalização Hippo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/complicações , Síndromes Mielodisplásicas/patologia
3.
Int J Mol Sci ; 24(9)2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37175742

RESUMO

Melanoma is considered a lethal and treatment-resistant skin cancer with a high risk of recurrence, making it a major clinical challenge. Our earlier studies documented that 1,25(OH)2D3 and its low-calcaemic analogues potentiate the effectiveness of dacarbazine and cediranib, a pan-VEGFR inhibitor. In the current study, a set of patient-derived melanoma cultures was established and characterised as a preclinical model of human melanoma. Thus, patient-derived cells were preconditioned with 1,25(OH)2D3 and treated with cediranib or vemurafenib, a BRAF inhibitor, depending on the BRAF mutation status of the patients enrolled in the study. 1,25(OH)2D3 preconditioning exacerbated the inhibition of patient-derived melanoma cell growth and motility in comparison to monotherapy with cediranib. A significant decrease in mitochondrial respiration parameters, such as non-mitochondrial oxygen consumption, basal respiration and ATP-linked respiration, was observed. It seems that 1,25(OH)2D3 preconditioning enhanced cediranib efficacy via the modulation of mitochondrial bioenergetics. Additionally, 1,25(OH)2D3 also decreased the viability and mobility of the BRAF+ patient-derived cells treated with vemurafenib. Interestingly, regardless of the strict selection, cancer-derived fibroblasts (CAFs) became the major fraction of cultured cells over time, suggesting that melanoma growth is dependent on CAFs. In conclusion, the results of our study strongly emphasise that the active form of vitamin D, 1,25(OH)2D3, might be considered as an adjuvant agent in the treatment of malignant melanoma.


Assuntos
Antineoplásicos , Melanoma , Humanos , Vitamina D/uso terapêutico , Vemurafenib/farmacologia , Vemurafenib/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/genética , Melanoma/tratamento farmacológico , Melanoma/patologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Vitaminas/uso terapêutico
4.
Mar Drugs ; 20(10)2022 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-36286442

RESUMO

Melanoma cells are highly invasive and metastatic tumor cells and commonly express molecular alterations that contribute to multidrug resistance (e.g., BRAFV600E mutation). Conventional treatment is not effective in a long term, requiring an exhaustive search for new alternatives. Recently, carotenoids from microalgae have been investigated as adjuvant in antimelanoma therapy due to their safety and acceptable clinical tolerability. Many of them are currently used as food supplements. In this review, we have compiled several studies that show microalgal carotenoids inhibit cell proliferation, cell migration and invasion, as well as induced cell cycle arrest and apoptosis in various melanoma cell lines. MAPK and NF-ĸB pathway, MMP and apoptotic factors are frequently affected after exposure to microalgal carotenoids. Fucoxanthin, astaxanthin and zeaxanthin are the main carotenoids investigated, in both in vitro and in vivo experimental models. Preclinical data indicate these compounds exhibit direct antimelanoma effect but are also capable of restoring melanoma cells sensitivity to conventional chemotherapy (e.g., vemurafenib and dacarbazine).


Assuntos
Antineoplásicos , Melanoma , Microalgas , Humanos , Vemurafenib/farmacologia , Vemurafenib/uso terapêutico , Microalgas/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas B-raf/uso terapêutico , Carotenoides/farmacologia , Carotenoides/uso terapêutico , Zeaxantinas/farmacologia , NF-kappa B , Melanoma/patologia , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Proliferação de Células , Mutação , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral
5.
Cells ; 11(7)2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35406796

RESUMO

High-dose ascorbate paradoxically acts as a pro-oxidant causing the formation of hydrogen peroxide in an oxygen dependent manner. Tumor cells (in particular melanoma cells) show an increased vulnerability to ascorbate induced reactive oxygen species (ROS). Therefore, high-dose ascorbate is a promising pharmacological approach to treating refractory melanomas, e.g., with secondary resistance to targeted BRAF inhibitor therapy. BRAF mutated melanoma cells were treated with ascorbate alone or in combination with the BRAF inhibitor vemurafenib. Viability, cell cycle, ROS production, and the protein levels of phospho-ERK1/2, GLUT-1 and HIF-1α were analyzed. To investigate the treatment in vivo, C57BL/6NCrl mice were subcutaneously injected with D4M.3A (BrafV600E) melanoma cells and treated with intraperitoneal injections of ascorbate with or without vemurafenib. BRAF mutated melanoma cell lines either sensitive or resistant to vemurafenib were susceptible to the induction of cell death by pharmacological ascorbate. Treatment of BrafV600E melanoma bearing mice with ascorbate resulted in plasma levels in the pharmacologically active range and significantly improved the therapeutic effect of vemurafenib. We conclude that intravenous high-dose ascorbate will be beneficial for melanoma patients by interfering with the tumor's energy metabolism and can be safely combined with standard melanoma therapies such as BRAF inhibitors without pharmacological interference.


Assuntos
Antineoplásicos , Melanoma , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Humanos , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/genética , Espécies Reativas de Oxigênio/uso terapêutico , Vemurafenib/farmacologia , Vemurafenib/uso terapêutico
6.
Lima; IETSI; dic. 2021.
Não convencional em Espanhol | BRISA | ID: biblio-1357647

RESUMO

INTRODUCCIÓN: El melanoma maligno (MM), es un tipo de cáncer de piel agresivo que afecta a los melanocitos. El MM representa solo el 4 % de los tumores malignos de la piel, pero es el responsable del 80 % de las muertes por estos tumores. Actualmente, la quimioterapia se usa con menos frecuencia que la inmunoterapia o la terapia dirigida. La inmunoterapia inhibe la proteína PD-1; y la terapia dirigida inhibe la proteína mutada BRAF y las proteínas quinasas MEK. A diferencia de la inmunoterapia (que puede usarse para todos los pacientes con MM metastásico), la terapia dirigida está indicada solo para aquellos pacientes con la mutación BRAF V600. La mutación BRAF está presente en el 50 % de los casos de MM y el 70 % de las mutaciones BRAF son de tipo V600E. En EsSalud, los pacientes adultos con MM avanzado o metastásico, con mutación BRAF V600E, inoperable y con ECOG 0-1 disponen de la inmunoterapia (nivolumab) y la quimioterapia como tratamientos de primera línea. No obstante, los especialistas en oncología señalan que la terapia dirigida podría ser más eficaz, en aquellos con la mutación BRAF V600E, que las opciones disponibles en EsSalud. De esta forma, sugieren el uso de vemurafenib más cobimetinib (V+C) como tratamiento de primera línea para estos pacientes. OBJETIVO: El objetivo del presente dictamen fue evaluar la eficacia y seguridad de V+C, comparado con nivolumab o quimioterapia, para el tratamiento de pacientes adultos con MM avanzado o metastásico, con mutación BRAF V600E, inoperable y con ECOG 0-1. METODOLOGÍA: Tras la búsqueda de la evidencia, se incluyeron cinco guías de práctica clínica (GPC) elaboradas por la National Comprehensive Cancer Network (NCCN), la American Society of Clinical Oncology (ASCO), el Cancer Council Australia (CCA), la European Society for Medical Oncology (ESMO) y la Scottish Intercollegiate Guidelines Network (SIGN); tres evaluaciones de tecnologías sanitarias (ETS) realizadas por el National Institute for Health and Care Excellence (NICE), la Canadian Agency for Drugs and Technologies in Health (CADTH) y el Institute for Quality and Efficiency in Healthcare (IQWiG); y dos revisiones sistemáticas con metaanálisis en red (RSMAR) desarrolladas por Zoratti et al. 2019 y Pike et al. 2017. RESULTADOS: Se llevó a cabo una búsqueda de evidencia científica con respecto al uso de V+C como tratamiento de primera línea en pacientes adultos con MM avanzado o metastásico, con mutación BRAF V600E, inoperable y con ECOG 0-1. CONCLUSIONES: El presente dictamen preliminar tuvo por objetivo evaluar la mejor evidencia disponible sobre la eficacia y seguridad de V+C, comparado con nivolumab o quimioterapia, para el tratamiento de pacientes adultos con MM avanzado o metastásico, con mutación BRAF, inoperable y con ECOG 0-1. Los desenlaces de interés fueron SG, calidad de vida y eventos adversos. Tras la búsqueda de la literatura se identificaron cinco GPC elaboradas por NCCN, CCA, ASCO, ESMO y SIGN; tres ETS desarrolladas por NICE, CADTH e IQWiG. No se identificaron ECA o estudios observacionales comparativos que respondieran a la pregunta PICO. Por tal motivo, se optó por incluir dos estudios que evaluaron de manera indirecta (metaanálisis en red) V+C vs. nivolumab o quimioterapia (la RSMAR de Zoratti et al. 2019 y la RSMAR de Pike et al. 2017). Cuatro GPC (NCCN, ASCO, ESMO, CCA) de las cinco evaluadas recomiendan nivolumab o V+C; la guía restante (SIGN), solo nivolumab; y ninguna guia mencionó a la quimioterapia como tratamiento de primera línea. La ESMO recomienda a los inhibidores de PD-1(nivolumab) como el tratamiento estándar de primera línea para los pacientes con MM metastásico, además, la CCA y la ESMO señalaron que es preferible iniciar con anti PD-1 (nivolumab); sobre todo en pacientes con buen estado de salud y cuya enfermedad no progrese rápidamente. Las ETS fueron disimiles en sus recomendaciones. La CADTH condicionó su recomendación a la reducción en el precio del medicamento; IQWiG concluyó que existe beneficio adicional a favor de V+C; pero que la información sobre eventos adversos (EA) presenta limitaciones importantes. Por su parte NICE no recomendó el uso de V+C por no ser costo-efectivo. Es de destacar que las tres ETS no incluyen entre sus comparadores a nivolumab o quimioterapia, medicamentos que son alternativas disponibles y con las que se tienen experiencia de uso en EsSalud. Las RSMAR sugieren que V+C es similar a nivolumab en términos de SG y EA. Aunque sugieren diferencias a favor de V+C, en comparación con quimioterapia, las estimaciones indirectas no cumplieron con el criterio de transitividad y presentaron limitaciones que afectan su validez. Además, reportes más recientes muestran una alta incidencia de EA serios para V+C diferentes a los reportados por las RSMAR. Dado que la evidencia disponible no ha mostrado que la relación riesgo-beneficio sea favorable a V+C, en comparación con nivolumab o quimioterapia, la aprobación de uso de V+C en EsSalud no sería una decisión costo-oportuna; tomando en cuenta la disponibilidad del tratamiento con nivolumab, el cual fue aprobado al ser comparado con quimioterapia, eficaz y seguro, recomendado por guías internacionales, con el cual se tiene experiencia de uso y es menos costoso. Por lo expuesto, el IETSI no aprueba el uso de vemurafenib más cobimetinib para el tratamiento de pacientes adultos con melanoma maligno avanzado o metastásico, con mutación BRAF, inoperable y con ECOG 0-1.


Assuntos
Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/uso terapêutico , Vemurafenib/uso terapêutico , Melanoma/complicações , Melanoma/tratamento farmacológico , Metástase Neoplásica , Eficácia , Análise Custo-Benefício , Combinação de Medicamentos
7.
Endocrine ; 67(1): 117-123, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31377969

RESUMO

PURPOSE: Anaplastic thyroid cancer (ATC) is rare but with poor prognosis. TRAIL can selectively induce apoptosis in cancer cells; however, resistance is quite common. Aim of our study was to evaluate TRAIL-induced apoptosis in ATC-derived cell lines, in vitro and in vivo, and the effect of combination with tyrosine kinase inhibitors (TKIs) selective for BRAF (vemurafenib) or Akt (MK-2206). METHODS: Four ATC-derived cell lines were used: C643, CAL62, HTh7, with activating mutation of RAS and copy gain of PI3K (HTh7) and, 8505C with activating mutation of BRAF. Cells were treated with TRAIL alone or in combination with vemurafenib or MK-2206. The pro-apoptotic effect of TRAIL alone or combined with TKIs was, also, evaluated in two mouse xenograft models (HTh7 and 8505C). RESULTS: C643, CAL62, and HTh7 cells were sensitive to TRAIL-induced apoptosis, whereas 8505C cells were resistant. Both in vitro and in vivo vemurafenib was able to increase the TRAIL-induced apoptosis in 8505C cells causing a slower tumor growth in 8505C xenograft compared to placebo, while MK-2206 did not have any additive effect on TRAIL treatment in HTh7 model. CONCLUSIONS: TRAIL is a promising therapeutic agent in ATC and in case of resistance vemurafenib may be a valid complementary therapy.


Assuntos
Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide , Animais , Apoptose , Linhagem Celular Tumoral , Camundongos , Ligante Indutor de Apoptose Relacionado a TNF , Carcinoma Anaplásico da Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Vemurafenib/farmacologia , Vemurafenib/uso terapêutico
8.
Dermatol Clin ; 37(2): 159-168, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30850038

RESUMO

Melanoma is rapidly evolving because of advances in noninvasive diagnosis, targeted therapies, and improved prognostic methods. This article discusses what is new in melanoma risk factors, prevention, clinical management, and targeted treatment. The incidence continues to increase worldwide, whereas mortality is steadily improving. This trend reinforces the importance of dermatologists comprehensively understanding all aspects of melanoma. Further research is needed to continue making a material impact on outcomes for patients.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Procedimentos Cirúrgicos Dermatológicos , Melanoma/terapia , Anti-Inflamatórios não Esteroides/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Biópsia , Café , Espectroscopia Dielétrica , Detecção Precoce de Câncer , Predisposição Genética para Doença , Genômica , Humanos , Doenças Inflamatórias Intestinais/epidemiologia , Ipilimumab/uso terapêutico , Melanoma/diagnóstico , Melanoma/epidemiologia , Melanoma/genética , Microscopia Confocal , Nivolumabe/uso terapêutico , Inibidores da Fosfodiesterase 5/uso terapêutico , Prevenção Primária , Prognóstico , Fatores de Proteção , Fatores de Risco , Prevenção Secundária , Biópsia de Linfonodo Sentinela , Fator de Proteção Solar , Protetores Solares/uso terapêutico , Ultrassonografia , Vemurafenib/uso terapêutico
9.
CNS Oncol ; 7(2): CNS10, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29708404

RESUMO

Pleomorphic xanthoastrocytomas can be very resistant to treatment if they progress after standard therapy with surgery and radiation. We present the case of a patient with a multiply recurrent pleomorphic xanthoastrocytoma which demonstrated a sustained partial response to a combination regimen of the BRAF inhibitor vemurafenib and tumor treating fields. The regimen proved tolerable and efficacious in this case.


Assuntos
Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Vemurafenib/uso terapêutico , Antineoplásicos/uso terapêutico , Astrocitoma/diagnóstico por imagem , Astrocitoma/genética , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Terapia Combinada , Terapia por Estimulação Elétrica/métodos , Evolução Fatal , Feminino , Humanos , Proteínas Proto-Oncogênicas B-raf/genética , Adulto Jovem
10.
Rev. Hosp. Ital. B. Aires (2004) ; 36(3): 84-90, sept. 2016. ilus
Artigo em Espanhol | LILACS | ID: biblio-1146685

RESUMO

El melanoma ha experimentado un aumento constante en su tasa de incidencia en las últimas cinco décadas a nivel mundial. El pronóstico del paciente con melanoma se relaciona con el estadio de la enfermedad al momento del diagnóstico, con una sobrevida global media de 6,2 meses en pacientes con melanoma metastásico. El avance en las investigaciones sobre la biología y el comportamiento tumoral permitió el desarrollo de nuevas terapias con distintos mecanismos de acción y mayor eficacia. En esta revisión se abordan las terapias biológicas en melanoma metastásico, su mecanismo de acción y principales resultados en ensayos clínicos. (AU)


Melanoma has experienced a consistent increase in incidence over the past five decades worldwide. The prognosis of patients with melanoma is related to the stage of disease at diagnosis, with a median overall survival of 6.2 months in metastatic melanoma. Progress in research on tumor biology allowed the development of new therapies with different mechanisms of action and greater efficiency. In this review, biologic therapies in metastatic melanoma, its mechanism of action and main results in clinical trials are discussed. (AU)


Assuntos
Humanos , Terapia Biológica , Melanoma/terapia , Metástase Neoplásica/terapia , Incidência , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Dacarbazina/efeitos adversos , Dacarbazina/uso terapêutico , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Ipilimumab/efeitos adversos , Ipilimumab/uso terapêutico , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/uso terapêutico , Vemurafenib/efeitos adversos , Vemurafenib/uso terapêutico , Nivolumabe/efeitos adversos , Nivolumabe/uso terapêutico , Imunoterapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA