Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Pathol ; 255(1): 72-83, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34124783

RESUMEN

Chordomas are primary bone tumors that arise in the cranial base, mobile spine, and sacrococcygeal region, affecting patients of all ages. Currently, there are no approved agents for chordoma patients. Here, we evaluated the anti-tumor efficacy of small molecule inhibitors that target oncogenic pathways in chordoma, as single agents and in combination, to identify novel therapeutic approaches with the greatest translational potential. A panel of small molecule compounds was screened in vivo against patient-derived xenograft (PDX) models of chordoma, and potentially synergistic combinations were further evaluated using chordoma cell lines and xenograft models. Among the tested agents, inhibitors of EGFR (BIBX 1382, erlotinib, and afatinib), c-MET (crizotinib), and mTOR (AZD8055) significantly inhibited tumor growth in vivo but did not induce tumor regression. Co-inhibition of EGFR and c-MET using erlotinib and crizotinib synergistically reduced cell viability in chordoma cell lines but did not result in enhanced in vivo activity. Co-inhibition of EGFR and mTOR pathways using afatinib and AZD8055 synergistically reduced cell viability in chordoma cell lines. Importantly, this dual inhibition completely suppressed tumor growth in vivo, showing improved tumor control. Together, these data demonstrate that individual inhibitors of EGFR, c-MET, and mTOR pathways suppress chordoma growth both in vitro and in vivo. mTOR inhibition increased the efficacy of EGFR inhibition on chordoma growth in several preclinical models. The insights gained from our study potentially provide a novel combination therapeutic strategy for patients with chordoma. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Afatinib/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cordoma/patología , Morfolinas/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Neurooncol ; 140(3): 529-538, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30414098

RESUMEN

PURPOSE: Meningiomas are a frequent tumor of the central nervous system. Although mostly benign, approximately 5% present as atypical or malignant tumors. Treatments for atypical meningiomas include gross total resection and radiotherapy, but about 33% of patients have recurrent tumors, sometimes as a higher grade. Recently, the brain penetrant anthelmintic drug, mebendazole, has shown promise as an anticancer agent in rodent models of glioblastoma and medulloblastoma. METHODS: The half maximal inhibitory concentration (IC50) effect on colony formation, cell proliferation, and caspase-3/7 markers of apoptosis of mebendazole with and without radiation was measured in vitro. Mice intracranially implanted with KT21MG1 human meningioma were administered mebendazole alone or in combination with radiation. Survival benefit was evaluated, while tumors were investigated by immunohistochemical staining for apoptosis, cell proliferation, and vascular density. RESULTS: In vitro experiments on meningioma cell lines showed the IC50 for mebendazole in the range of 0.26-0.42 µM. Mebendazole alone induced cytotoxicity, however the combination had a greater reduction in colony formation and resulted in higher levels of cleaved caspase-3. The in vivo study showed both, mebendazole alone and the combination, to have a survival benefit with an increase in apoptosis, and decreases in tumor cell and vascular proliferation. CONCLUSION: These preclinical findings indicate that mebendazole alone or in combination with radiation can be considered for the treatment of malignant meningioma. The mechanism of action for this combination may include an increase in apoptosis, a reduction in proliferation and angiogenesis, or a combination of these effects.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Mebendazol/administración & dosificación , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/radioterapia , Meningioma/tratamiento farmacológico , Meningioma/radioterapia , Animales , Antihelmínticos/administración & dosificación , Antineoplásicos/administración & dosificación , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Concentración 50 Inhibidora , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo
3.
Southeast Asian J Trop Med Public Health ; 45(6): 1264-70, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26466412

RESUMEN

Mebendazole (MBZ) is an anthelmintic drug which inhibits tubulin polymerization and eventually causes apoptosis in target organisms. Antitumor activity of MBZ has been reported in various cancers. The aim of this study was to investigate the effect of MBZ on cholangiocarcinoma (CCA) cells in vitro and in vivo. MBZ reduced cell proliferation in the KKU-M213 cell line associated with a remarkable enhancement of caspase-3 gene expression and enzyme activity. Oral administration of MBZ slightly reduced the growth rate of subcutaneously xeno-grafted KKU-M213 in nude mice. The TUNEL assay showed an increase of apoptotic cell numbers in the xenograft tumor tissue of MBZ-treated mice. The data obtained in this study suggested that MBZ can suppress CCA cell proliferation via caspase-3 activated apoptosis. Further investigation of the antitumor effects of MBZ might support the use of MBZ as an alternative drug for CCA treatment.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/patología , Mebendazol/farmacología , Animales , Neoplasias de los Conductos Biliares , Conductos Biliares Intrahepáticos , Línea Celular Tumoral , Ratones
4.
Pharm Res ; 29(7): 1832-42, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22354837

RESUMEN

PURPOSE: Physiologically based models, when verified in pre-clinical species, optimally predict human pharmacokinetics. However, modeling of intestinal metabolism has been a gap. To establish in vitro/in vivo scaling factors for metabolism, the expression and activity of CYP enzymes were characterized in the intestine and liver of beagle dog. METHODS: Microsomal protein abundance in dog tissues was determined using testosterone-6ß-hydroxylation and 7-hydroxycoumarin-glucuronidation as markers for microsomal protein recovery. Expressions of 7 CYP enzymes were estimated based on quantification of proteotypic tryptic peptides using multiple reaction monitoring mass spectrometry. CYP3A12 and CYP2B11 activity was evaluated using selective marker reactions. RESULTS: The geometric mean of total microsomal protein was 51 mg/g in liver and 13 mg/cm in intestine, without significant differences between intestinal segments. CYP3A12, followed by CYP2B11, were the most abundant CYP enzymes in intestine. Abundance and activity were higher in liver than intestine and declined from small intestine to colon. CONCLUSIONS: CYP expression in dog liver and intestine was characterized, providing a basis for in vitro/in vivo scaling of intestinal and hepatic metabolism.


Asunto(s)
Sistema Enzimático del Citocromo P-450/análisis , Intestinos/enzimología , Hígado/enzimología , Microsomas/enzimología , Secuencia de Aminoácidos , Animales , Hidrocarburo de Aril Hidroxilasas/análisis , Hidrocarburo de Aril Hidroxilasas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Familia 2 del Citocromo P450 , Perros , Intestinos/química , Hígado/química , Espectrometría de Masas , Microsomas/química , Datos de Secuencia Molecular , Esteroide Hidroxilasas/análisis , Esteroide Hidroxilasas/metabolismo
5.
Oncotarget ; 12(14): 1326-1338, 2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34262644

RESUMEN

The five-year survival rate for metastatic pancreatic cancer is currently only 3%, which increases to 13% with local invasion only and to 39% with localized disease at diagnosis. Here we evaluated repurposed mebendazole, an approved anthelminthic drug, to determine how mebendazole might work at the different stages of pancreatic cancer formation and progression. We asked if mebendazole could prevent initiation of pancreatic intraepithelial neoplasia precursor lesions, interfere with stromal desmoplasia, or suppress tumor growth and liver metastasis. In both the Kras LSL.G12D/+; Pdx1-Cre (KC) mouse model of caerulein-induced inflammatory pancreatitis and the Kras LSL.G12D/+; Tp53 R172H/+; Pdx1-Cre (KPC) mouse model of advanced pancreatic cancer, mebendazole significantly reduced pancreas weight, dysplasia and intraepithelial neoplasia formation, compared to controls. Mebendazole significantly reduced trichrome-positive fibrotic connective tissue and α-SMA-positive activated pancreatic stellate cells that heralds fibrogenesis. In the aggressive KPC model, mebendazole significantly suppressed pancreatic tumor growth, both as an early and late intervention. Mebendazole reduced the overall incidence of pancreatic cancer and severity of liver metastasis in KPC mice. Using early models of pancreatic cancer, treatment with mebendazole resulted in less inflammation, decreased dysplasia, with the later stage model additionally showing a decreased tumor burden, less advanced tumors, and a reduction of metastasis. We conclude that mebendazole should be investigated further as a component of adjuvant therapy to slow progression and prevent metastasis, and well as for primary prevention in the highest risk patients.

6.
Endocr Relat Cancer ; 27(3): 123-136, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31846433

RESUMEN

The most common thyroid malignancy is papillary thyroid cancer. While a majority respond to therapy and have a favorable prognosis, some papillary thyroid cancers persist. This subset may dedifferentiate to anaplastic thyroid cancer, an aggressive, highly invasive and rapidly fatal cancer. Thyroid cancer patients at risk for disease progression and metastasis need earlier, safer and more effective therapies. The purpose of this translational study was to determine if mebendazole could be repurposed to effectively treat thyroid cancer, in particular before metastasis. In vitro, mebendazole potently inhibited the growth of a panel of human papillary and anaplastic thyroid cancer cells. In papillary (B-CPAP) and anaplastic (8505c) cell lines, mebendazole increased the percentage of cells in G2/M cell cycle arrest and induced late stage apoptosis by activation of the caspase-3 pathway. In aggressive 8505c cells, mebendazole significantly repressed migratory and invasive potential in a wound healing and transwell invasion assay and inhibited expression of phosphorylated Akt and Stat3 and reduced Gli1. In vivo, mebendazole treatment resulted in significant orthotopic thyroid tumor regression (B-CPAP) and growth arrest (8505c), with treated tumors displaying reduced expression of the proliferation maker KI67 and less vascular epithelium as indicated by CD31+ immunohistochemistry. Most importantly, daily oral mebendazole prevented established thyroid tumors from metastasizing to the lung. Given the low toxicity and published anticancer mechanisms of mebendazole, this novel preclinical study of mebendazole in thyroid cancer has promising therapeutic implications for patients with treatment refractory papillary or anaplastic thyroid cancer.


Asunto(s)
Neoplasias Pulmonares/secundario , Mebendazol/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias Pulmonares/prevención & control , Ratones , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/irrigación sanguínea , Neoplasias de la Tiroides/patología
7.
SLAS Technol ; 24(1): 28-40, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30289729

RESUMEN

Glioblastoma (GBM) is a lethal brain cancer with a median survival time of approximately 15 months following treatment. Common in vitro GBM models for drug screening are adherent and do not recapitulate the features of human GBM in vivo. Here we report the genomic characterization of nine patient-derived, spheroid GBM cell lines that recapitulate human GBM characteristics in orthotopic xenograft models. Genomic sequencing revealed that the spheroid lines contain alterations in GBM driver genes such as PTEN, CDKN2A, and NF1. Two spheroid cell lines, JHH-136 and JHH-520, were utilized in a high-throughput drug screen for cell viability using a 1912-member compound library. Drug mechanisms that were cytotoxic in both cell lines were Hsp90 and proteasome inhibitors. JHH-136 was uniquely sensitive to topoisomerase 1 inhibitors, while JHH-520 was uniquely sensitive to Mek inhibitors. Drug combination screening revealed that PI3 kinase inhibitors combined with Mek or proteasome inhibitors were synergistic. However, animal studies to test these drug combinations in vivo revealed that Mek inhibition alone was superior to the combination treatments. These data show that these GBM spheroid lines are amenable to high-throughput drug screening and that this dataset may deliver promising therapeutic leads for future GBM preclinical studies.


Asunto(s)
Antineoplásicos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Glioblastoma/patología , Mutación , Esferoides Celulares/efectos de los fármacos , Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Células Tumorales Cultivadas
8.
Nat Commun ; 9(1): 5410, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30575736

RESUMEN

Olfactory neuroblastoma (ONB) is a rare malignant neoplasm arising in the upper portion of the sinonasal cavity. To better understand the genetic bases for ONB, here we perform whole exome and whole genome sequencing as well as single nucleotide polymorphism array analyses in a series of ONB patient samples. Deletions involving the dystrophin (DMD) locus are found in 12 of 14 (86%) tumors. Interestingly, one of the remaining tumors has a deletion in LAMA2, bringing the number of ONBs with deletions of genes involved in the development of muscular dystrophies to 13 or 93%. This high prevalence implicates an unexpected functional role for genes causing hereditary muscular dystrophies in ONB.

9.
Oncotarget ; 7(42): 68571-68584, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27612418

RESUMEN

Inheritance of a gene mutation leads to the initiation of 5 to 10% of most cancers, including colon cancer cases. We developed a chemoprevention strategy using a novel combination of the non-steroidal anti-inflammatory (NSAID) sulindac plus the anthelminthic benzimidazole, mebendazole. This oral drug combination was effective in the ApcMin/+ mouse model of Familial Adenomatous Polyposis (FAP). Treatment with 35 mg/kg daily mebendazole reduced the number of intestinal adenomas by 56% (P = 0.0002), 160 ppm sulindac by 74% (P < 0.0001), and the combination by 90% (P < 0.0001). The combination significantly reduced microadenomas, polyp number and size in both the small intestines and colon when compared to untreated controls or sulindac alone. Mebendazole as a single agent decreased COX2 expression, blood vessel formation, VEGFR2 phosphorylation, and worked synergistically with sulindac to reduce overexpression of MYC, BCL2, and various pro-inflammatory cytokines. Given the low toxicity of mebendazole, these preclinical findings support the consideration of clinical trials for high risk cancer patients using mebendazole either alone or in combination. The findings have implications for populations with moderate and above risk for developing cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Colon/efectos de los fármacos , Neoplasias del Colon/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/prevención & control , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Línea Celular Tumoral , Colon/patología , Neoplasias del Colon/patología , Células HCT116 , Células HT29 , Humanos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/patología , Masculino , Mebendazol/administración & dosificación , Ratones Endogámicos C57BL , Ratones Desnudos , Sulindac/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA