Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 27(14): 3096-3100, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28545975

RESUMEN

RAD51 is a vital component of the homologous recombination DNA repair pathway and is overexpressed in drug-resistant cancers, including aggressive triple negative breast cancer (TNBC). A proposed strategy for improving therapeutic outcomes for patients is through small molecule inhibition of RAD51, thereby sensitizing tumor cells to DNA damaging irradiation and/or chemotherapy. Here we report structure-activity relationships for a library of quinazolinone derivatives. A novel RAD51 inhibitor (17) displays up to 15-fold enhanced inhibition of cell growth in a panel of TNBC cell lines compared to compound B02, and approximately 2-fold increased inhibition of irradiation-induced RAD51 foci formation. Additionally, compound 17 significantly inhibits TNBC cell sensitivity to DNA damage, implying a potentially targeted therapy for cancer treatment.


Asunto(s)
Antineoplásicos/química , Inhibidores Enzimáticos/química , Quinazolinonas/química , Recombinasa Rad51/antagonistas & inhibidores , Antineoplásicos/farmacología , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cinamatos/síntesis química , Cinamatos/química , Cinamatos/toxicidad , Daño del ADN/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Quinazolinas/síntesis química , Quinazolinas/química , Quinazolinas/toxicidad , Quinazolinonas/farmacología , Recombinasa Rad51/metabolismo , Relación Estructura-Actividad , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
3.
NAR Cancer ; 3(2): zcab022, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34316709

RESUMEN

Chemotherapy is used as a standard-of-care against cancers that display high levels of inherent genome instability. Chemotherapy induces DNA damage and intensifies pressure on the DNA repair pathways that can lead to deregulation. There is an urgent clinical need to be able to track the emergence of DNA repair driven chemotherapy resistance and tailor patient staging appropriately. There have been numerous studies into chemoresistance but to date no study has elucidated in detail the roles of the key DNA repair components in resistance associated with the frontline clinical combination of anthracyclines and taxanes together. In this study, we hypothesized that the emergence of chemotherapy resistance in triple negative breast cancer was driven by changes in functional signaling in the DNA repair pathways. We identified that consistent pressure on the non-homologous end joining pathway in the presence of genome instability causes failure of the key kinase DNA-PK, loss of p53 and compensation by p73. In-turn a switch to reliance on the homologous recombination pathway and RAD51 recombinase occurred to repair residual double strand DNA breaks. Further we demonstrate that RAD51 is an actionable target for resensitization to chemotherapy in resistant cells with a matched gene expression profile of resistance highlighted by homologous recombination in clinical samples.

4.
Front Mol Biosci ; 7: 134, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32766277

RESUMEN

The emergence of clinical resistance in repeatedly treated cancers extends from the primary tumor's capability to exploit genome instability to adapt, escape, and progress. Triple negative breast cancer serves as a good example of such a response demonstrating poor clinical outcome due to a high rate of cellular heterogeneity resulting in metastatic relapse. The capability to effectively track the emergence of therapeutic resistance in real-time and adapt the clinical response is the holy grail for precision medicine and has yet to be realized. In this review we present liquid biopsy using CTCs and ctDNA as a potential replacement and/or addition to the current diagnostic tests to deliver personalized therapies to patients with advanced breast cancer. We outline current uses of liquid biopsy in the metastatic breast cancer setting and discuss their limitations. In addition, we provide a detailed overview of common genome instability events in patients with metastatic breast cancer and how these can be tracked using liquid biopsy.

5.
Cancer Treat Rev ; 41(1): 35-45, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25467108

RESUMEN

The DNA damage response (DDR) is essential for maintaining genomic stability and cell survival. However, when tumour cells with deficiencies in HR are faced with radio- and chemotherapies they are forced to rely on error-prone, alternative repair pathways or aberrant HR for survival; threatening genome integrity and driving further mutation. Accurate therapeutic targeting of the key drivers of DNA repair can circumvent survival pathways and avoid aggressive therapy resistant mutants. Several studies have identified that stabilization of the cancer genome in HR deficient cells can be achieved by overexpression of the recombinase RAD51. Radio- and chemotherapeutic resistance is associated with overactive HR repair mechanisms. However no clinical trials have directly targeted RAD51, despite RAD51 displaying synergy in several drug screens against multiple cancer types. Currently synthetic lethality targeting the DDR pathways and HR deficiency has had clinical success with BRCA1 functional loss and PARP inhibition. In this review we suggest that clinical outcomes could be improved by additionally targeting RAD51. We examine the latest developments in directly and indirectly targeting RAD51. We scrutinize the potential treatment efficacy and future clinical applications of RAD51 inhibitors as single agents and in combination with other therapies and consider the best therapeutic options.


Asunto(s)
Reparación del ADN/genética , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Recombinasa Rad51/antagonistas & inhibidores , Daño del ADN , Recombinación Homóloga , Humanos , Neoplasias/genética
6.
Mol Cancer Ther ; 14(10): 2321-31, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26294743

RESUMEN

The triple-negative breast cancer (TNBC) subtype represents a cancer that is highly aggressive with poor patient outcome. Current preclinical success has been gained through synthetic lethality, targeting genome instability with PARP inhibition in breast cancer cells that harbor silencing of the homologous recombination (HR) pathway. Histone deacetylase inhibitors (HDACi) are a class of drugs that mediate epigenetic changes in expression of HR pathway genes. Here, we compare the activity of the pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA), the class I/IIa HDAC inhibitor valproic acid (VPA), and the HDAC1/2-specific inhibitor romidepsin (ROMI) for their capability to regulate DNA damage repair gene expression and in sensitizing TNBC to PARPi. We found that two of the HDACis tested, SAHA and ROMI, but not VPA, indeed inhibit HR repair and that RAD51, BARD1, and FANCD2 represent key proteins whose inhibition is required for HDACi-mediated therapy with PARP inhibition in TNBC. We also observed that restoration of BRCA1 function stabilizes the genome compared with mutant BRCA1 that results in enhanced polyploid population after combination treatment with HDACi and PARPi. Furthermore, we found that overexpression of the key HR protein RAD51 represents a mechanism for this resistance, promoting aberrant repair and the enhanced polyploidy observed. These findings highlight the key components of HR in guiding synthetic lethality with PARP inhibition and support the rationale for utilizing the novel combination of HDACi and PARPi against TNBC in the clinical setting.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA1/genética , Bencimidazoles/farmacología , Expresión Génica/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , Enzimas Reparadoras del ADN/genética , Depsipéptidos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Epigénesis Genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Humanos , Ácidos Hidroxámicos/farmacología , Concentración 50 Inhibidora , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ácido Valproico/farmacología , Vorinostat
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA