Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NPJ Precis Oncol ; 6(1): 81, 2022 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-36323843

RESUMEN

Inactivating mutations of MBD4 have been reported in subsets of various tumors. A deficiency of this DNA glycosylase, recognizing specifically T:G mismatch resulting from the deamination of methyl-cytosine, results in a hypermutated phenotype due to the accumulation of CpG>TpG transitions. Here, we hypothesize that the difference in DNA metabolism consecutive to MBD4 deficiency may result in specific cytotoxicities in MBD4-deficient tumor cells in a synthetic lethality fashion. After a large-scale drug repurposing screen, we show in two isogenic MBD4 knock-out cell models that the inactivation of MBD4 sensitizes cancer cells to cytidine analogs. We further confirm the exquisite activity of gemcitabine in an MBD4-deficient co-clinical model as (i) it completely prevented the development of an MBD4-deficient uveal melanoma patient-derived xenograft and (ii) treatment in the corresponding patient resulted in an exceptional tumor response. These data suggest that patients harboring MBD4-deficient tumors may be treated efficiently by cytidine analogs.

2.
EMBO Mol Med ; 14(9): e15670, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-36069081

RESUMEN

Centrosome amplification, the presence of more than two centrosomes in a cell is a common feature of most human cancer cell lines. However, little is known about centrosome numbers in human cancers and whether amplification or other numerical aberrations are frequently present. To address this question, we have analyzed a large cohort of primary human epithelial ovarian cancers (EOCs) from 100 patients. We found that rigorous quantitation of centrosome number in tumor samples was extremely challenging due to tumor heterogeneity and extensive tissue disorganization. Interestingly, even if centrosome clusters could be identified, the incidence of centrosome amplification was not comparable to what has been described in cultured cancer cells. Surprisingly, centrosome loss events where a few or many nuclei were not associated with centrosomes were clearly noticed and overall more frequent than centrosome amplification. Our findings highlight the difficulty of characterizing centrosome numbers in human tumors, while revealing a novel paradigm of centrosome number defects in EOCs.


Asunto(s)
Centrosoma , Neoplasias Ováricas , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular , Centrosoma/metabolismo , Centrosoma/patología , Femenino , Humanos , Neoplasias Ováricas/patología
3.
Eur Radiol ; 27(10): 4435-4444, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28374075

RESUMEN

OBJECTIVE: This study aimed to explore the antitumour effect of the DNA repair inhibitor, DT01 (the cholesterol conjugated form of Dbait), as an adjunct treatment to enhance the therapeutic efficacy of transarterial chemoembolization (TACE) in pre-clinical models of hepatocellular carcinoma (HCC). METHODS: A rabbit model bearing liver tumours was either left untreated or treated with TACE or with a combination of TACE+DT01. Tumour growth was monitored by ultrasound. These results were further confirmed in mice grafted with an intrahepatic human HCC model treated with doxorubicin (DOX) alone or DOX+DT01. RESULTS: The combination of DT01 with TACE in a rabbit liver model led to a significant decrease in tumour volume (p=0.03). Colour Doppler and immunohistochemical staining revealed a strong decrease in vascularization in the DT01+TACE-treated group preventing the tumour growth restart observed after TACE alone. Similarly, the DT01 combination with DOX led to significant anti-tumour efficacy compared to DOX alone (p=0.02) in the human HCC model. In addition, a significant decrease in vascularization in the group receiving combination DT01 and DOX treatment was observed. CONCLUSIONS: DT01 is well tolerated and may potentiate HCC treatment by enhancing the DNA-damaging and anti-vascularization effect of TACE with doxorubicin. KEY POINTS: • DT01 combined with TACE leads to significant anti-tumour efficacy without additional toxicity. • A potential anti-angiogenic role of DT01 was identified in preclinical models. • DT01 may potentiate HCC treatment by enhancing the efficacy of TACE.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica , Colesterol/análogos & derivados , Reparación del ADN/efectos de los fármacos , ADN/uso terapéutico , Doxorrubicina/uso terapéutico , Neoplasias Hepáticas/terapia , Animales , Carcinoma Hepatocelular/genética , Quimioembolización Terapéutica/métodos , Colesterol/uso terapéutico , Daño del ADN , Modelos Animales de Enfermedad , Humanos , Neoplasias Hepáticas/genética , Masculino , Conejos , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
4.
Oncotarget ; 7(11): 12927-36, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26887045

RESUMEN

Radiolabelled melanin ligands offer an interesting strategy for the treatment of disseminated pigmented melanoma. One of these molecules, ICF01012 labelled with iodine 131, induced a significant slowing of melanoma growth. Here, we have explored the combination of [131I]ICF01012 with coDbait, a DNA repair inhibitor, to overcome melanoma radioresistance and increase targeted radionuclide therapy (TRT) efficacy. In human SK-Mel 3 melanoma xenograft, the addition of coDbait had a synergistic effect on tumor growth and median survival. The anti-tumor effect was additive in murine syngeneic B16Bl6 model whereas coDbait combination with [131I]ICF01012 did not increase TRT side effects in secondary pigmented tissues (e.g. hair follicles, eyes). Our results confirm that DNA lesions induced by TRT were not enhanced with coDbait association but, the presence of micronuclei and cell cycle blockade in tumor shows that coDbait acts by interrupting or delaying DNA repair. In this study, we demonstrate for the first time, the usefulness of DNA repair traps in the context of targeted radionuclide therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Reparación del ADN/efectos de los fármacos , ADN/farmacología , Melanoma Experimental/tratamiento farmacológico , Animales , Sinergismo Farmacológico , Femenino , Humanos , Radioisótopos de Yodo/farmacología , Masculino , Melanoma/patología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Quinoxalinas/farmacología , Proteína Tumoral Controlada Traslacionalmente 1 , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Cancer Ther ; 15(1): 15-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26637369

RESUMEN

Metastatic liver disease from colorectal cancer is a significant clinical problem. This is mainly attributed to nonresectable metastases that frequently display low sensitivities to available chemotherapies and develop drug resistance partly via hyperactivation of some DNA repair functions. Combined therapies have shown some disease control; however, there is still a need for more efficient chemotherapies to achieve eradication of colorectal cancer liver metastasis. We investigated the tolerance and efficacy of a novel class of DNA repair inhibitors, Dbait, in association with conventional chemotherapy. Dbait mimics double-strand breaks and activates damage signaling, consequently inhibiting single- and double-stranded DNA repair enzyme recruitment. In vitro, Dbait treatment increases sensitivity of HT29 and HCT116 colorectal cancer cell lines. In vivo, the pharmacokinetics, biodistribution and the efficacy of the cholesterol-conjugated clinical form of Dbait, DT01, were assessed. The chemosensitizing abilities of DT01 were evaluated in association with oxaliplatin and 5-fluorouracil in intrahepatic HT29 xenografted mice used as a model for colorectal cancer liver metastasis. The high uptake of DT01 indicates that the liver is a specific target. We demonstrate significant antitumor efficacy in a liver metastasis model with DT01 treatment in combination with oxaliplatin and 5-fluorouracil (mean: 501 vs. 872 mm(2), P = 0.02) compared to chemotherapy alone. The decrease in tumor volume is further associated with significant histologic changes in necrosis, proliferation, angiogenesis and apoptosis. Repeated cycles of DT01 do not increase chemotherapy toxicity. Combining DT01 with conventional chemotherapy may prove to be a safe and effective therapeutic strategy in the treatment of metastatic liver cancer.


Asunto(s)
Antineoplásicos/farmacología , Colesterol/análogos & derivados , Neoplasias Colorrectales/patología , Reparación del ADN/efectos de los fármacos , ADN/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Hepáticas/secundario , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Colesterol/administración & dosificación , Colesterol/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , ADN/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Fluorouracilo/farmacología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/secundario , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
PLoS One ; 7(7): e40567, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815765

RESUMEN

BACKGROUND: Glioma is the most aggressive tumor of the brain and the most efficient treatments are based on radiotherapy. However, tumors are often resistant to radiotherapy due to an enhanced DNA repair activity. Short and stabilized DNA molecules (Dbait) have recently been proposed as an efficient strategy to inhibit DNA repair in tumor. METHODOLOGY/PRINCIPAL FINDINGS: The distribution of three formulations of Dbait, (i) Dbait alone, (ii) Dbait associated with polyethylenimine, and (iii) Dbait linked with cholesterol (coDbait), was evaluated one day after intratumoral delivery in an RG2 rat glioma model. Dbait molecule distribution was assessed in the whole organ with 2D-FRI and in brain sections. CoDbait was chosen for further studies given its good retention in the brain, cellular localization, and efficacy in inducing the activation of DNA repair effectors. The radiosensitizing effect of coDbait was studied in four groups of rats bearing RG2-glioma: no treatment, radiotherapy only, coDbait alone, and CoDbait with radiotherapy. Treatment started 7 days after tumor inoculation and consisted of two series of treatment in two weeks: coDbait injection followed by a selective 6-Gy irradiation of the head. We evaluated the radiosensitizing effect using animal survival, tumor volume, cell proliferation, and vasculature characteristics with multiparametric MRI. CoDbait with radiotherapy improved the survival of rats bearing RG2-glioma by reducing tumor growth and cell proliferation without altering tumor vasculature. CONCLUSION/SIGNIFICANCE: coDbait is therefore a promising molecular therapy to sensitize glioma to radiotherapy.


Asunto(s)
Colesterol/metabolismo , ADN/metabolismo , ADN/farmacología , Glioblastoma/patología , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Transporte Biológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Química Farmacéutica , ADN/efectos adversos , ADN/química , Roturas del ADN de Doble Cadena , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glioblastoma/irrigación sanguínea , Glioblastoma/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/efectos de la radiación , Imagen por Resonancia Magnética , Masculino , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/efectos de la radiación , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Neostriado/patología , Neostriado/efectos de la radiación , Neovascularización Patológica , Polietileneimina/química , Fármacos Sensibilizantes a Radiaciones/efectos adversos , Fármacos Sensibilizantes a Radiaciones/química , Ratas , Análisis de Supervivencia , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación
7.
J Biol Chem ; 287(12): 8803-15, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22270370

RESUMEN

DNA damage triggers a complex signaling cascade involving a multitude of phosphorylation events. We found that the threonine 7 (Thr-7) residue of heat shock protein 90α (Hsp90α) was phosphorylated immediately after DNA damage. The phosphorylated Hsp90α then accumulated at sites of DNA double strand breaks and formed repair foci with slow kinetics, matching the repair kinetics of complex DNA damage. The phosphorylation of Hsp90α was dependent on phosphatidylinositol 3-kinase-like kinases, including the DNA-dependent protein kinase (DNA-PK) in particular. DNA-PK plays an essential role in the repair of DNA double strand breaks by nonhomologous end-joining and in the signaling of DNA damage. It is also present in the cytoplasm of the cell and has been suggested to play a role in cytoplasmic signaling pathways. Using stabilized double-stranded DNA molecules to activate DNA-PK, we showed that an active DNA-PK complex could be assembled in the cytoplasm, resulting in phosphorylation of the cytoplasmic pool of Hsp90α. In vivo, reverse phase protein array data for tumors revealed that basal levels of Thr-7-phosphorylated Hsp90α were correlated with phosphorylated histone H2AX levels. The Thr-7 phosphorylation of the ubiquitously produced and secreted Hsp90α may therefore serve as a surrogate biomarker of DNA damage. These findings shed light on the interplay between central DNA repair enzymes and an essential molecular chaperone.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas HSP90 de Choque Térmico/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular Tumoral , Femenino , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/genética , Humanos , Ratones , Ratones Desnudos , Fosforilación , Ratas
8.
J Gastroenterol ; 47(3): 266-75, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22068457

RESUMEN

BACKGROUND: Dbait molecules are a new class of DNA repair inhibitors triggering false DNA damage signaling in cancer cells. Dbait has already been shown to be effective in combination with radiotherapy. The aim of this study was to assess the adjuvant impact of Dbait on chemotherapy in vitro and in mouse models of colorectal cancer. METHODS: We assessed DNA repair efficiency over time, in vitro, in human colon adenocarcinoma HT-29 (wild-type KRAS) and HCT-116 (mutated KRAS) cell lines treated with Dbait in combination with 5-fluorouracil and/or camptothecin. Genetically engineered mice spontaneously developing colorectal tumors in the intestines were selected for the evaluation of treatment efficacy. RESULTS: Dbait delayed the repair of DNA damage induced by chemotherapy in vitro. In APC (+/1638N) mutant mice, the combination of Dbait and chemotherapy decreased tumor size more effectively than chemotherapy alone (median size: 3.6 vs. 10.85 mm(2), P < 0.05). In APC (+/1638N)/KRAS ( V12G ) mutant mice, animals treated with a combination of Dbait and chemotherapy survived significantly longer than animals treated by chemotherapy alone (median survival: 210 vs. 194 days, P < 0.05). A quarter of all the animals treated by chemotherapy alone died as rapidly as untreated animals, whereas the first death was delayed by 29 days by the addition of Dbait. No increase in toxicity due to Dbait was observed in either mouse model. CONCLUSIONS: The use of Dbait to inhibit DNA repair may be an effective additional treatment for increasing the efficacy of chemotherapy in colon or rectal cancer, independently of KRAS status.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Reparación del ADN/efectos de los fármacos , Adenocarcinoma/patología , Animales , Camptotecina/administración & dosificación , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Daño del ADN/efectos de los fármacos , Fluorouracilo/administración & dosificación , Ingeniería Genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Tiempo , Proteínas ras/genética
9.
Surg Endosc ; 26(3): 847-52, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22042585

RESUMEN

BACKGROUND: Peritoneal carcinomatosis is an unmet medical need. Laparoscopy offers a unique opportunity to control and to steer the operating environment during surgery by loading carbon dioxide with a therapeutic substance and creating the so-called therapeutic capnoperitoneum. We have treated a human sample of peritoneal carcinomatosis from an endometrial adenocarcinoma ex vivo just after surgery. METHODS: A nontoxic therapeutic agent (Dbait) was aerosolized into a box containing diseased human peritoneum under a pressure of 12 mmHg CO(2). Dbait (noncoding DNA fragments) acts through jamming DNA damage sensing and signaling, ultimately inhibiting DNA repair system of cancer cells. Dbait were coupled to cholesterol molecules to facilitate intracellular uptake, and to Cyanine (Cy5) to allow detection by fluorescence. In a control experiment, the same solution was applied to the other half of the sample using conventional lavage. RESULTS: Physical results revealed fluorescence within the tumor up to 1 mm depth in the therapeutic capnoperitoneum sample and no uptake in the lavage sample. Biological results showed intranuclear phosphorylation of H2AX in the nebulized sample and no activity in the lavage sample. Importantly, tumor nodules showed more activity than the neighbor, normal peritoneum. Detection of histone gamma-H2AX (phosphorylated H2AX) reveals activation of DNA-dependent protein kinase (DNA-PK) by Dbait, which has been shown to be the key step for sensitization to genotoxic therapy. CONCLUSIONS: Dbait are taken up by cancer cells and have a biological activity up to 1 mm depth. Nebulization of the molecule is significantly more effective than conventional lavage. This proof of principle supports the need for clinical studies applying therapeutic capnoperitoneum together with Dbait for treating peritoneal carcinomatosis.


Asunto(s)
Antineoplásicos/administración & dosificación , Dióxido de Carbono/administración & dosificación , Carcinoma/terapia , Neoplasias Peritoneales/terapia , Neumoperitoneo Artificial/métodos , ARN Interferente Pequeño/administración & dosificación , Adenocarcinoma/terapia , Adulto , Aerosoles , Carbocianinas , Terapia Combinada , Neoplasias Endometriales/terapia , Diseño de Equipo , Femenino , Fluorescencia , Colorantes Fluorescentes , Histonas/metabolismo , Humanos , Laparoscopía/métodos
10.
Development ; 138(18): 3943-54, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21862558

RESUMEN

We aim to evaluate environmental and genetic effects on the expansion/proliferation of committed single cells during embryonic development, using melanoblasts as a paradigm to model this phenomenon. Melanoblasts are a specific type of cell that display extensive cellular proliferation during development. However, the events controlling melanoblast expansion are still poorly understood due to insufficient knowledge concerning their number and distribution in the various skin compartments. We show that melanoblast expansion is tightly controlled both spatially and temporally, with little variation between embryos. We established a mathematical model reflecting the main cellular mechanisms involved in melanoblast expansion, including proliferation and migration from the dermis to epidermis. In association with biological information, the model allows the calculation of doubling times for melanoblasts, revealing that dermal and epidermal melanoblasts have short but different doubling times. Moreover, the number of trunk founder melanoblasts at E8.5 was estimated to be 16, a population impossible to count by classical biological approaches. We also assessed the importance of the genetic background by studying gain- and loss-of-function ß-catenin mutants in the melanocyte lineage. We found that any alteration of ß-catenin activity, whether positive or negative, reduced both dermal and epidermal melanoblast proliferation. Finally, we determined that the pool of dermal melanoblasts remains constant in wild-type and mutant embryos during development, implying that specific control mechanisms associated with cell division ensure half of the cells at each cell division to migrate from the dermis to the epidermis. Modeling melanoblast expansion revealed novel links between cell division, cell localization within the embryo and appropriate feedback control through ß-catenin.


Asunto(s)
Diferenciación Celular , Crecimiento y Desarrollo/fisiología , Melanocitos/fisiología , Modelos Biológicos , Modelos Teóricos , Animales , Animales Recién Nacidos , Proliferación Celular , Células Cultivadas , Dermis/citología , Dermis/embriología , Embrión de Mamíferos , Células Epidérmicas , Epidermis/embriología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
11.
Clin Cancer Res ; 15(4): 1308-16, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19190126

RESUMEN

PURPOSE: Enhanced DNA repair activity is often associated with tumor resistance to radiotherapy. We hypothesized that inhibiting DNA damage repair would sensitize tumors to radiation-induced DNA damage. EXPERIMENTAL DESIGN: A novel strategy for inhibiting DNA repair was tested. We designed small DNA molecules that mimic DNA double-strand breaks (called Dbait) and act by disorganizing damage signaling and DNA repair. We analyzed the effects of Dbait in cultured cells and on xenografted tumors growth and performed preliminary studies of their mechanism(s) of action. RESULTS: The selected Dbait molecules activate H2AX phosphorylation in cell culture and in xenografted tumors. In vitro, this activation correlates with the reduction of Nijmegen breakage syndrome 1 and p53-binding protein 1 repair foci formation after irradiation. Cells are sensitized to irradiation and do not efficiently repair DNA damage. In vivo, Dbait induces regression of radioresistant head and neck squamous cell carcinoma (Hep2) and melanoma (SK28 and LU1205) tumors. The combination of Dbait32Hc treatment and fractionated radiotherapy significantly enhanced the therapeutic effect. Tumor growth control by Dbait molecules depended directly on the dose and was observed with various irradiation protocols. The induction of H2AX phosphorylation in tumors treated with Dbait suggests that it acts in vivo through the induction of "false" DNA damage signaling and repair inhibition. CONCLUSIONS: These data validate the concept of introducing small DNA molecules, which mimic DNA damage, to trigger "false" signaling of DNA damage and impair DNA repair of damaged chromosomes. This new strategy could provide a new method for enhancing radiotherapy efficiency in radioresistant tumors.


Asunto(s)
Daño del ADN , Reparación del ADN/efectos de los fármacos , Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Línea Celular Tumoral , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Femenino , Histonas/metabolismo , Humanos , Ratones , Fosforilación , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...