Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Eur J Med Chem ; 265: 116114, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38194775

RESUMEN

The BRCA2-RAD51 interaction remains an intriguing target for cancer drug discovery due to its vital role in DNA damage repair mechanisms, which cancer cells become particularly reliant on. Moreover, RAD51 has many synthetically lethal partners, including PARP1-2, which can be exploited to induce synthetic lethality in cancer. In this study, we established a 19F-NMR-fragment based approach to identify RAD51 binders, leading to two initial hits. A subsequent SAR program identified 46 as a low micromolar inhibitor of the BRCA2-RAD51 interaction. 46 was tested in different pancreatic cancer cell lines, to evaluate its ability to inhibit the homologous recombination DNA repair pathway, mediated by BRCA2-RAD51 and trigger synthetic lethality in combination with the PARP inhibitor talazoparib, through the induction of apoptosis. Moreover, we further analyzed the 46/talazoparib combination in 3D pancreatic cancer models. Overall, 46 showed its potential as a tool to evaluate the RAD51/PARP1-2 synthetic lethality mechanism, along with providing a prospect for further inhibitors development.


Asunto(s)
Antineoplásicos , Neoplasias Pancreáticas , Humanos , Antineoplásicos/química , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Línea Celular Tumoral , Reparación del ADN , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/metabolismo , Mutaciones Letales Sintéticas
2.
Nat Commun ; 12(1): 5966, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645815

RESUMEN

The BRCA2 tumor suppressor protects genome integrity by promoting homologous recombination-based repair of DNA breaks, stability of stalled DNA replication forks and DNA damage-induced cell cycle checkpoints. BRCA2 deficient cells display the radio-resistant DNA synthesis (RDS) phenotype, however the mechanism has remained elusive. Here we show that cells without BRCA2 are unable to sufficiently restrain DNA replication fork progression after DNA damage, and the underrestrained fork progression is due primarily to Primase-Polymerase (PRIMPOL)-mediated repriming of DNA synthesis downstream of lesions, leaving behind single-stranded DNA gaps. Moreover, we find that BRCA2 associates with the essential DNA replication factor MCM10 and this association suppresses PRIMPOL-mediated repriming and ssDNA gap formation, while having no impact on the stability of stalled replication forks. Our findings establish an important function for BRCA2, provide insights into replication fork control during the DNA damage response, and may have implications in tumor suppression and therapy response.


Asunto(s)
Proteína BRCA2/genética , ADN Primasa/genética , ADN de Neoplasias/genética , ADN de Cadena Simple/genética , ADN Polimerasa Dirigida por ADN/genética , Proteínas de Mantenimiento de Minicromosoma/genética , Enzimas Multifuncionales/genética , Reparación del ADN por Recombinación , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Primasa/antagonistas & inhibidores , ADN Primasa/metabolismo , Replicación del ADN , ADN de Neoplasias/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Proteínas de Mantenimiento de Minicromosoma/antagonistas & inhibidores , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Enzimas Multifuncionales/antagonistas & inhibidores , Enzimas Multifuncionales/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Cell Chem Biol ; 28(6): 835-847.e5, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33662256

RESUMEN

BRCA2 controls RAD51 recombinase during homologous DNA recombination (HDR) through eight evolutionarily conserved BRC repeats, which individually engage RAD51 via the motif Phe-x-x-Ala. Using structure-guided molecular design, templated on a monomeric thermostable chimera between human RAD51 and archaeal RadA, we identify CAM833, a 529 Da orthosteric inhibitor of RAD51:BRC with a Kd of 366 nM. The quinoline of CAM833 occupies a hotspot, the Phe-binding pocket on RAD51 and the methyl of the substituted α-methylbenzyl group occupies the Ala-binding pocket. In cells, CAM833 diminishes formation of damage-induced RAD51 nuclear foci; inhibits RAD51 molecular clustering, suppressing extended RAD51 filament assembly; potentiates cytotoxicity by ionizing radiation, augmenting 4N cell-cycle arrest and apoptotic cell death and works with poly-ADP ribose polymerase (PARP)1 inhibitors to suppress growth in BRCA2-wildtype cells. Thus, chemical inhibition of the protein-protein interaction between BRCA2 and RAD51 disrupts HDR and potentiates DNA damage-induced cell death, with implications for cancer therapy.


Asunto(s)
Proteína BRCA2/antagonistas & inhibidores , Recombinasa Rad51/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Muerte Celular/efectos de los fármacos , Cristalografía por Rayos X , Daño del ADN , Humanos , Modelos Moleculares , Conformación Molecular , Unión Proteica/efectos de los fármacos , Recombinasa Rad51/química , Recombinasa Rad51/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Células Tumorales Cultivadas
4.
Chembiochem ; 22(1): 129-133, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-32979005

RESUMEN

Ibrutinib is an FDA-approved drug to treat B-lymphoid malignancies, which functions mechanistically as a covalent inhibitor for Bruton's tyrosine kinase (BTK). During the course of screening more potent and selective BTK inhibitors, we discovered that MM2-48, an ibrutinib analogue that contains the alkynyl amide functional group in place of the acrylamide warhead, exhibits a much stronger cytotoxicity. Comparative chemoproteomic profiling of the targets of ibrutinib and MM2-48 revealed that the alkynyl amide warhead exhibits much higher reactivity in proteomes. Unexpectedly, MM2-48 covalently targets a functional cysteine in a BRCA2 and CDKN1A-interacting protein, BCCIP, and significantly inhibits DNA damage repair. Our findings suggest that simultaneous inhibition of BTK activity and DNA damage repair might be a more effective therapeutic strategy for combating B-cell malignancies.


Asunto(s)
Adenina/análogos & derivados , Proteína BRCA2/antagonistas & inhibidores , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteómica , Adenina/química , Adenina/farmacología , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Humanos , Estructura Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Piperidinas/química , Inhibidores de Proteínas Quinasas/química
5.
Int J Oncol ; 57(6): 1245-1261, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33174058

RESUMEN

Triple­negative breast cancer (TNBC) accounts for 10­15% of all breast cancer cases. TNBCs lack estrogen and progesterone receptors and express low levels of HER2, and therefore do not respond to hormonal or anti­HER2 therapies. TNBC is a particularly aggressive form of breast cancer that generally displays poorer prognosis compared to other breast cancer subtypes. TNBC is chemotherapy sensitive, and this treatment remains the standard of care despite its limited benefit. Recent advances with novel agents have been made for specific subgroups with PD­L1+ tumors or germline Brca­mutated tumors. However, only a fraction of these patients responds to immune checkpoint or PARP inhibitors and even those who do respond often develop resistance and relapse. Various new agents and combination strategies have been explored to further understand molecular and immunological aspects of TNBC. In this review, we discuss clinical trials in the management of TNBC as well as perspectives for potential future treatments.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mastectomía , Recurrencia Local de Neoplasia/epidemiología , Neoplasias de la Mama Triple Negativas/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Mama/patología , Mama/cirugía , Quimioterapia Adyuvante/métodos , Resistencia a Antineoplásicos/genética , Femenino , Mutación de Línea Germinal , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/prevención & control , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Pronóstico , Supervivencia sin Progresión , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/mortalidad
6.
Anticancer Agents Med Chem ; 19(2): 206-212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30417796

RESUMEN

BACKGROUND: Poly-ADP-ribosylation, that is, adding ADP-ribose moieties to a protein, is a unique type of protein post-translational modification that regulates various cellular processes such as DNA repair, mitosis, transcription, and cell growth. Small-molecule inhibitors of poly-ADP-ribose polymerase 1 (PARP1) have been developed as anticancer agents because inhibition of PARP enzymes may be a synthetic lethal strategy for cancers with or BRCA2 mutations. However, there are still questions surrounding PARP inhibitors. METHODS/RESULTS: Data were collected from Pubmed, Medline, through searching of these keywords: "PARP", "BRCA", "Synthetic lethal" and "Tankyrase inhibitors". We describe the current knowledge of PARP inhibition and its effects on DNA damage; mechanisms of resistance to PARP inhibitors; the evolution of PARP inhibitors; and the potential use of PARP5a/b (tankyrases) inhibitors in cancer treatment. CONCLUSION: PARP inhibitors are already showing promise as therapeutic tools, especially in the management of BRCA-mutated breast and ovarian cancers but also in tumors with dysfunctional BRCA genes. Small-molecule tankyrase inhibitors are important for increasing our understanding of tankyrase biology.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/química , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Mutación , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Bibliotecas de Moléculas Pequeñas/química
8.
Endocr Relat Cancer ; 25(1): 69-82, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29066501

RESUMEN

Clinical implications of the BRCA2 expression level on treatments of ovarian cancer are controversial. Here, we demonstrated that platinum-resistant cancer had a higher percentage of high BRCA2 level (87.5% vs 43.6%, P = 0.001), and that patients with a low BRCA2 level in cancer tissues had longer progression-free survival (with a median time of 28.0 vs 12.0 months, P < 0.001) and platinum-free duration (with a median time of 19.0 vs 5.0 months, P < 0.001) compared with those with a high BRCA2 level. In human ovarian cancer cell lines CAOV-3 and ES-2, cisplatin induced an upregulation of the RAD51 protein, which was inhibited after silencing BRCA2; silencing BRCA2 enhanced the action of cisplatin in vitro and in vivo Knockdown of BRCA2 promoted cisplatin-induced autophagy. Interestingly, the autophagy blocker chloroquine enhanced cisplatin in BRCA2-silenced cells accompanied by an increase in apoptotic cells, which did not occur in BRCA2-intact cells; chloroquine enhanced the efficacy of cisplatin against BRCA2-silenced CAOV-3 tumors in vivo, with an increase in LC3-II level in tumor tissues. Sensitization of cisplatin was also observed in BRCA2-silenced CAOV-3 cells after inhibiting ATG7, confirming that chloroquine modulated the sensitivity via the autophagy pathway. These data suggest that a low BRCA2 level can predict better platinum sensitivity and prognosis, and that the modulation of autophagy can be a chemosensitizer for certain cancers.


Asunto(s)
Adenocarcinoma de Células Claras/patología , Antineoplásicos/farmacología , Autofagia , Proteína BRCA2/antagonistas & inhibidores , Cisplatino/farmacología , Cistadenocarcinoma Seroso/patología , Neoplasias Ováricas/patología , Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma de Células Claras/metabolismo , Apoptosis/efectos de los fármacos , Proteína BRCA2/metabolismo , Biomarcadores de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/metabolismo , Resistencia a Antineoplásicos , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
ACS Chem Biol ; 12(10): 2491-2497, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-28841282

RESUMEN

In BRCA2-defective cells, poly(adenosine diphosphate [ADP]-ribose) polymerase inhibitors can trigger synthetic lethality, as two independent DNA-repairing mechanisms are simultaneously impaired. Here, we have pharmacologically induced synthetic lethality, which was triggered by combining two different small organic molecules. When administered with a BRCA2-Rad51 disruptor in nonmutant cells, Olaparib showed anticancer activity comparable to that shown when administered alone in BRCA2-defective cells. This strategy could represent an innovative approach to anticancer drug discovery and could be extended to other synthetic lethality pathways.


Asunto(s)
Proteína BRCA2/antagonistas & inhibidores , Ftalazinas/farmacología , Piperazinas/farmacología , Recombinasa Rad51/antagonistas & inhibidores , Antineoplásicos/química , Antineoplásicos/farmacología , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Línea Celular Tumoral , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Modelos Moleculares , Mutación , Ftalazinas/química , Piperazinas/química , Conformación Proteica , Recombinasa Rad51/metabolismo
10.
Nat Commun ; 8: 13905, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28067217

RESUMEN

The CRISPR-Cas9 system provides a versatile toolkit for genome engineering that can introduce various DNA lesions at specific genomic locations. However, a better understanding of the nature of these lesions and the repair pathways engaged is critical to realizing the full potential of this technology. Here we characterize the different lesions arising from each Cas9 variant and the resulting repair pathway engagement. We demonstrate that the presence and polarity of the overhang structure is a critical determinant of double-strand break repair pathway choice. Similarly, single nicks deriving from different Cas9 variants differentially activate repair: D10A but not N863A-induced nicks are repaired by homologous recombination. Finally, we demonstrate that homologous recombination is required for repairing lesions using double-stranded, but not single-stranded DNA as a template. This detailed characterization of repair pathway choice in response to CRISPR-Cas9 enables a more deterministic approach for designing research and therapeutic genome engineering strategies.


Asunto(s)
Proteína BRCA2/genética , Sistemas CRISPR-Cas , ADN/genética , Edición Génica/métodos , Genoma Humano , Recombinasa Rad51/genética , Reparación del ADN por Recombinación , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteína 9 Asociada a CRISPR , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN/metabolismo , Roturas del ADN de Doble Cadena , Endonucleasas/genética , Endonucleasas/metabolismo , Células HEK293 , Humanos , Células K562 , Osteoblastos/citología , Osteoblastos/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/metabolismo
11.
Nucleic Acids Res ; 44(12): 5743-57, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27131361

RESUMEN

DNA double-strand breaks (DSBs) are toxic lesions, which if improperly repaired can result in cell death or genomic instability. DSB repair is usually facilitated by the classical non-homologous end joining (C-NHEJ), or homologous recombination (HR) pathways. However, a mutagenic alternative NHEJ pathway, microhomology-mediated end joining (MMEJ), can also be deployed. While MMEJ is suppressed by C-NHEJ, the relationship between HR and MMEJ is less clear. Here, we describe a role for HR genes in suppressing MMEJ in human cells. By monitoring DSB mis-repair using a sensitive HPRT assay, we found that depletion of HR proteins, including BRCA2, BRCA1 or RPA, resulted in a distinct mutational signature associated with significant increases in break-induced mutation frequencies, deletion lengths and the annealing of short regions of microhomology (2-6 bp) across the break-site. This signature was dependent on CtIP, MRE11, POLQ and PARP, and thus indicative of MMEJ. In contrast to CtIP or MRE11, depletion of BRCA1 resulted in increased partial resection and MMEJ, thus revealing a functional distinction between these early acting HR factors. Together these findings indicate that HR factors suppress mutagenic MMEJ following DSB resection.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN/metabolismo , Reparación del ADN por Recombinación , Proteína de Replicación A/genética , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/metabolismo , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Secuencia de Bases , Bioensayo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Endodesoxirribonucleasas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Proteína Homóloga de MRE11 , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína de Replicación A/antagonistas & inhibidores , Proteína de Replicación A/metabolismo , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , ADN Polimerasa theta
12.
Oncotarget ; 7(15): 20825-39, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26959114

RESUMEN

Human tumor heterogeneity promotes therapeutic failure by increasing the likelihood of resistant cell subpopulations. The PARP-1 inhibitor olaparib is approved for use in BRCA-mutated ovarian cancers but BRCA2-reversion mutations lead to functional homologous recombination repair (HRR) and olaparib resistance. To overcome that resistance and expand use of PARP1 inhibition to cancers with functional HRR, we developed an antisense strategy to render the majority of tumor cells in a population BRCA2-deficient. We predicted that this strategy would render HRR-proficient tumor cells sensitive to olaparib and prevent emergence of resistance in a tumor cell population heterogeneous for HRR proficiency. We report that BRCA2 downregulation sensitized multiple human tumor cell lines (but not non-cancer human kidney cells) to olaparib and, combined with olaparib, increased aneuploidy and chromosomal translocations in human tumor cells. In a mixed HRR-proficient and HRR-deficient cell population, olaparib monotherapy allowed outgrowth of HRR-proficient cells resistant to subsequent olaparib treatment. Combined BRCA2 inhibition and olaparib treatment prevented selection of HRR-proficient cells and inhibited proliferation of the entire population. Treatment with BRCA2 siRNA and olaparib decreased ovarian xenograft growth in mice more effectively than either treatment alone. In vivo use of BRCA2 antisense oligonucleotides may be a viable option to expand clinical use of olaparib and prevent resistance.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA2/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/farmacología , Piperazinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína BRCA2/genética , Biomarcadores de Tumor/genética , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , ARN Interferente Pequeño/genética , Reparación del ADN por Recombinación/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Prog Drug Res ; 71: 181-3, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26939289

RESUMEN

The focus of molecular docking is to computationally simulate the molecular recognition process. A binding interaction between a small molecule ligand and protein may result in activation or inhibition of the protein. The docking method using BRCA1 or BRCA2 genes and COX proteins is carefully texted in this chapter to check docking of the best inhibitor molecule.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA2/antagonistas & inhibidores , Bioprospección/métodos , Inhibidores de la Ciclooxigenasa/farmacología , Descubrimiento de Drogas/métodos , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Fitoquímicos/farmacología , Extractos Vegetales/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/metabolismo , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Proteína BRCA2/química , Proteína BRCA2/metabolismo , Sitios de Unión , Ciclooxigenasa 1/química , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa/química , Inhibidores de la Ciclooxigenasa/metabolismo , Bases de Datos de Proteínas , Humanos , Terapia Molecular Dirigida , Neoplasias/enzimología , Fitoquímicos/química , Fitoquímicos/metabolismo , Fitoterapia , Extractos Vegetales/química , Extractos Vegetales/metabolismo , Plantas Medicinales , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
14.
J Med Chem ; 59(1): 335-57, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26652717

RESUMEN

We discovered and developed a novel series of tetrahydropyridophthlazinones as poly(ADP-ribose) polymerase (PARP) 1 and 2 inhibitors. Lead optimization led to the identification of (8S,9R)-47 (talazoparib; BMN 673; (8S,9R)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-2,7,8,9-tetrahydro-3H-pyrido[4,3,2-de]phthalazin-3-one). The novel stereospecific dual chiral-center-embedded structure of this compound has enabled extensive and unique binding interactions with PARP1/2 proteins. (8S,9R)-47 demonstrates excellent potency, inhibiting PARP1 and PARP2 enzyme activity with Ki = 1.2 and 0.87 nM, respectively. It inhibits PARP-mediated PARylation in a whole-cell assay with an EC50 of 2.51 nM and prevents proliferation of cancer cells carrying mutant BRCA1/2, with EC50 = 0.3 nM (MX-1) and 5 nM (Capan-1), respectively. (8S,9R)-47 is orally available, displaying favorable pharmacokinetic (PK) properties and remarkable antitumor efficacy in the BRCA1 mutant MX-1 breast cancer xenograft model following oral administration as a single-agent or in combination with chemotherapy agents such as temozolomide and cisplatin. (8S,9R)-47 has completed phase 1 clinical trial and is currently being studied in phase 2 and 3 clinical trials for the treatment of locally advanced and/or metastatic breast cancer with germline BRCA1/2 deleterious mutations.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Ftalazinas/síntesis química , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/síntesis química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Animales , Antineoplásicos/farmacocinética , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Proliferación Celular/efectos de los fármacos , Ensayos Clínicos Fase I como Asunto , Descubrimiento de Drogas , Femenino , Mutación de Línea Germinal , Humanos , Cinética , Ratones , Ratones Desnudos , Modelos Moleculares , Ftalazinas/farmacocinética , Poli(ADP-Ribosa) Polimerasa-1 , Unión Proteica , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Am Chem Soc ; 137(17): 5741-7, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25849366

RESUMEN

(-)-Lomaiviticin A (1) and the monomeric lomaiviticin aglycon [aka: (-)-MK7-206, (3)] are cytotoxic agents that induce double-strand breaks (DSBs) in DNA. Here we elucidate the cellular responses to these agents and identify synthetic lethal interactions with specific DNA repair factors. Toward this end, we first characterized the kinetics of DNA damage by 1 and 3 in human chronic myelogenous leukemia (K562) cells. DSBs are rapidly induced by 3, reaching a maximum at 15 min post addition and are resolved within 4 h. By comparison, DSB production by 1 requires 2-4 h to achieve maximal values and >8 h to achieve resolution. As evidenced by an alkaline comet unwinding assay, 3 induces extensive DNA damage, suggesting that the observed DSBs arise from closely spaced single-strand breaks (SSBs). Both 1 and 3 induce ataxia telangiectasia mutated- (ATM-) and DNA-dependent protein kinase- (DNA-PK-) dependent production of phospho-SER139-histone H2AX (γH2AX) and generation of p53 binding protein 1 (53BP1) foci in K562 cells within 1 h of exposure, which is indicative of activation of nonhomologous end joining (NHEJ) and homologous recombination (HR) repair. Both compounds also lead to ataxia telangiectasia and Rad3-related- (ATR-) dependent production of γH2AX at later time points (6 h post addition), which is indicative of replication stress. 3 is also shown to induce apoptosis. In accord with these data, 1 and 3 were found to be synthetic lethal with certain mutations in DNA DSB repair. 1 potently inhibits the growth of breast cancer type 2, early onset- (BRCA2-) deficient V79 Chinese hamster lung fibroblast cell line derivative (VC8), and phosphatase and tensin homologue deleted on chromosome ten- (PTEN-) deficient human glioblastoma (U251) cell lines, with LC50 values of 1.5 ± 0.5 and 2.0 ± 0.6 pM, respectively, and selectivities of >11.6 versus the isogenic cell lines transfected with and expressing functional BRCA2 and PTEN genes. 3 inhibits the growth of the same cell lines with LC50 values of 6.0 ± 0.5 and 11 ± 4 nM and selectivities of 84 and 5.1, for the BRCA2 and PTEN mutants, respectively. These data argue for the evaluation of these agents as treatments for tumors that are deficient in BRCA2 and PTEN, among other DSB repair factors.


Asunto(s)
Proteína BRCA2/antagonistas & inhibidores , Roturas del ADN de Doble Cadena/efectos de los fármacos , Fluorenos/farmacología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proteína BRCA2/deficiencia , Línea Celular , Cricetulus , Reparación del ADN , Fluorenos/química , Humanos , Células K562 , Cinética , Estructura Molecular , Fosfohidrolasa PTEN/deficiencia , Relación Estructura-Actividad
16.
Drugs ; 75(2): 231-40, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25616434

RESUMEN

Olaparib (Lynparza™) is an oral, small molecule, poly (ADP-ribose) polymerase inhibitor being developed by AstraZeneca for the treatment of solid tumours. The primary indication that olaparib is being developed for is BRCA mutation-positive ovarian cancer. A capsule formulation of the drug has received approval for use in this setting in the EU and USA, and a tablet formulation is in global phase III trials (including in the USA, EU, Australia, Brazil, Canada, China, Israel, Japan, Russia and South Korea). In addition, phase III trials in breast, gastric and pancreatic cancer are underway/planned, and phase I/II investigation is being conducted in other malignancies, including prostate cancer, non-small cell lung cancer, Ewing's sarcoma and advanced cancer. This article summarizes the milestones in the development of olaparib leading to this first approval for ovarian cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Aprobación de Drogas , Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/uso terapéutico , Piperazinas/uso terapéutico , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Ensayos Clínicos Fase III como Asunto , Femenino , Humanos , Estructura Molecular , Mutación , Neoplasias Ováricas/genética , Ftalazinas/administración & dosificación , Ftalazinas/farmacología , Piperazinas/administración & dosificación , Piperazinas/farmacología
17.
ChemMedChem ; 10(2): 296-303, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25470112

RESUMEN

The development of small molecules that inhibit protein-protein interactions continues to be a challenge in chemical biology and drug discovery. Herein we report the development of indole-based fragments that bind in a shallow surface pocket of a humanised surrogate of RAD51. RAD51 is an ATP-dependent recombinase that plays a key role in the repair of double-strand DNA breaks. It both self-associates, forming filament structures with DNA, and interacts with the BRCA2 protein through a common "FxxA" tetrapeptide motif. We elaborated previously identified fragment hits that target the FxxA motif site and developed small-molecule inhibitors that are approximately 500-fold more potent than the initial fragments. The lead compounds were shown to compete with the BRCA2-derived Ac-FHTA-NH2 peptide and the self-association peptide of RAD51, but they had no effect on ATP binding. This study is the first reported elaboration of small-molecular-weight fragments against this challenging target.


Asunto(s)
Proteína BRCA2/metabolismo , Recombinasa Rad51/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Secuencias de Aminoácidos , Proteína BRCA2/antagonistas & inhibidores , Sitios de Unión , Diseño de Fármacos , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Estructura Terciaria de Proteína , Pyrococcus furiosus/enzimología , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/genética , Bibliotecas de Moléculas Pequeñas/metabolismo , Relación Estructura-Actividad , Termodinámica
18.
Oncotarget ; 5(9): 2778-91, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24784564

RESUMEN

Indoleamine 2,3-dioxygenase-1 (IDO) is an immunosuppressive molecule expressed by most human tumors. IDO levels correlate with poor prognosis in cancer patients and IDO inhibitors are under investigation to enhance endogenous anticancer immunosurveillance. Little is known of immune-independent functions of IDO relevant to cancer therapy. We show, for the first time, that IDO mediates human tumor cell resistance to a PARP inhibitor (olaparib), gamma radiation, cisplatin, and combined treatment with olaparib and radiation, in the absence of immune cells. Antisense-mediated reduction of IDO, alone and (in a synthetic lethal approach) in combination with antisense to the DNA repair protein BRCA2 sensitizes human lung cancer cells to olaparib and cisplatin. Antisense reduction of IDO decreased NAD+ in human tumor cells. NAD+ is essential for PARP activity and these data suggest that IDO mediates treatment resistance independent of immunity and at least partially due to a previously unrecognized role for IDO in DNA repair. Furthermore, IDO levels correlated with accumulation of tumor cells in G1 and depletion of cells in G2/M of the cell cycle, suggesting that IDO effects on cell cycle may also modulate sensitivity to radiation and chemotherapeutic agents. IDO is a potentially valuable therapeutic target in cancer treatment, independent of immune function and in combination with other therapies.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Rayos gamma , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Tolerancia a Radiación , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Western Blotting , Ciclo Celular , Proliferación Celular , Cisplatino/administración & dosificación , Terapia Combinada , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neoplasias/patología , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
19.
Mol Cancer ; 13: 94, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24775809

RESUMEN

It is well known that the activation of Aurora A/B (Aur A/B) or inactivation of BRCA1/2 induces tumor formation. Others and we have reported that the mutual suppression between Aur A/B and BRCA1/2 may manipulate cancer cell growth and tumorigenesis, however, the interactive regulation and mechanism between these molecules are still elusive. In this study, by consecutive silencing of Aur A/B or/and BRCA1/2 with specific shRNAs, we showed that, in BRCA2-deficient pancreatic cancer cell line Capan-1 and in ovarian cancer cell line OVCA433, Aur A/B and BRCA1/2 inversely regulated the expression of each other likely through proteasome-mediated proteolysis but not through gene transcription. Aur A/B and BRCA1/2 conversely regulated cell cycle progression mainly through control of p53 and cyclin A. Moreover, the disruption of Aur A/B blocked abnormal cytokinesis and decreased cell multinuclearity and chromosome tetraploidy, whereas the deprivation of BRCA1/2 promoted the abnormal cytokinesis and enhanced the cell multinuclearity and tetraploidy. Furthermore, we showed by animal assays that the depletion of Aur A/B inhibited tumor growth of both cell lines, while the knockdown of BRCA1/2 promoted the tumor growth. However, the concurrent silencing of Aur A/B and BRCA1/2 diminished the effects of these molecules on the regulation of cell cycle, cytokinesis, and tetraploidy, leading to the burdened tumor sizes similar to those induced by scrambled shRNA-treated control cells. In summary, our study revealed that the negative interplay between Aur A/B and BRCA1/2 inversely controls the cell proliferation, cell cycle progression, cell multinuclearity, and tetraploidization to modulate tumorigenesis.


Asunto(s)
Aurora Quinasa A/genética , Aurora Quinasa B/genética , Proteína BRCA1/genética , Proteína BRCA2/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Neoplasias Pancreáticas/genética , Animales , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/metabolismo , Aurora Quinasa B/antagonistas & inhibidores , Aurora Quinasa B/metabolismo , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/metabolismo , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Ciclo Celular/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ploidias , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Curr Cancer Drug Targets ; 12(4): 329-38, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22385509

RESUMEN

Over the past two decades, progresses in colorectal cancer treatment have significantly improved patient survival and quality of life. However, unresectable metastatic colorectal cancer remains virtually incurable, making the search for new effective therapeutics mandatory. An important limitation to the development of new agents has been the difficulty to exploit mutated tumor suppressors or "undruggable" oncogenes as a target. Recently, evidence that mutations in tumor suppressors, such as BRCA1/2, make cancer cells highly susceptible to inhibitors of a compensatory DNA repair pathway [poly-(ADP-ribose) polymerase 1 (PARP1)] has broadened the range of possible therapeutic targets by extending it to gene products that are in a "synthetic lethal" relationship with oncogenes and tumor suppressors. Inhibition of such targets blocks specific buffer-mechanisms that are required for survival in the presence of defined oncogenic mutations, but not in their absence. As a consequence, selective elimination of mutation-bearing cells results. This approach has led to identify compounds that are highly active in the presence of different types of mutated tumor suppressors and oncogenes, including DNA repair genes, RAS, and Myc. In addition, ongoing studies promise to identify new mechanisms which, when pharmacologically interfered with, will selectively eradicate mutated cancer cells. Here, we revise and discuss these new aspects of cancer biology and highlight their potential applications in colorectal cancer treatment.


Asunto(s)
Adenoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Adenoma/genética , Adenoma/prevención & control , Animales , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/prevención & control , Reparación de la Incompatibilidad de ADN/efectos de los fármacos , Reparación de la Incompatibilidad de ADN/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Ratones , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras) , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/agonistas , Proteína p53 Supresora de Tumor/genética , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Proteínas ras/antagonistas & inhibidores , Proteínas ras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA