Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
An Acad Bras Cienc ; 94(1): e20191476, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35018996

RESUMEN

The aim of present study, to evaluate the genotoxic potential of 1-(4-(3,3-dimethyl-1,6-dioxo-2,3,4,6,11,13-hexahydro-1H-indazolo[1,2b] phthalazine-13yl)phenyl)-2-phenylazetidine-3-yl-acetate which was synthesised assuming that it may be a pharmaceutical raw material and found to inhibit human carbonic anhydrase I, II isozymes. To determine the genotoxic potential of this phthalazine substituted ß-lactam compound, chromosomal aberration (CA) and micronucleus (MN) tests were implemented in human peripheral blood lymphocytes. In these tests, lymphocyte cultures were treated with four concentrations (30, 15, 7.5, 3.75 µg/mL) of test compound and simultaneously with negative control (sterile distilled water), solvent control (DMSO) positive control (MMC). According to our results, CA frequencies were significantly increased in two high applied concentrations (30, 15 µg/mL) compared with negative and solvent control. MN frequencies were significantly increased in three applied concentrations (30, 15, 7.5 µg/mL) except lowest concentration (3.75 µg/mL) compared with solvent control. Mitotic indices were also affected by treatment with test compound. The obtained results provide evidence to demonstrate that new phthalazine substituted ß-lactam derivative can exert genotoxic and cytotoxic effects in peripheral human lymphocytes especially at high concentrations.


Asunto(s)
Micronúcleos con Defecto Cromosómico , beta-Lactamas , Células Cultivadas , Aberraciones Cromosómicas/inducido químicamente , Daño del ADN , Relación Dosis-Respuesta a Droga , Humanos , Linfocitos , Pruebas de Micronúcleos , Ftalazinas/toxicidad , beta-Lactamas/toxicidad
2.
ChemMedChem ; 16(23): 3600-3614, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34665510

RESUMEN

Leishmaniasis and Chagas diseases are two of the most important parasitic diseases in the world. Both belong to the category of Neglected Tropical Diseases, and they cannot be prevented by vaccination. Their treatments are founded in outdated drugs that possess many pernicious side-effects and they're not easy to administer. With the aim of discovering new compounds that could serve as anti-trypanosomal drugs, an antiparasitic study of a synthetic compound family has been conducted. A series of new 1,4-bis(alkylamino)- and 1-alkylamino-4-chloroazine and benzoazine derivatives 1-4 containing imidazole rings have been synthesized and identified. Their structures showed a possible interest based on previous work. Their in vitro anti-Leishmania infantum, anti-L. braziliensis, anti-L. donovani and anti-T. cruzi activity were tested, as well as the inhibition of Fe-SOD enzymes. It was found that some of them exhibited quite relevant values indicative of being worthy of future more detailed studies, as most of them showed activity to more than only one parasite species, especially compound 3 c was active for the three studied Leishmania species and also for T. cruzi, which is a very interesting trait as it covers a wide spectrum.


Asunto(s)
Imidazoles/farmacología , Ftalazinas/farmacología , Piridazinas/farmacología , Tripanocidas/farmacología , Animales , Chlorocebus aethiops , Imidazoles/síntesis química , Imidazoles/toxicidad , Leishmania braziliensis/efectos de los fármacos , Leishmania donovani/efectos de los fármacos , Leishmania infantum/efectos de los fármacos , Pruebas de Sensibilidad Parasitaria , Ftalazinas/síntesis química , Ftalazinas/toxicidad , Piridazinas/síntesis química , Piridazinas/toxicidad , Tripanocidas/síntesis química , Tripanocidas/toxicidad , Trypanosoma cruzi/efectos de los fármacos , Células Vero
3.
Nucleic Acids Res ; 49(18): 10493-10506, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34551432

RESUMEN

The antitumor activity of poly(ADP-ribose) polymerase inhibitors (PARPis) has been ascribed to PARP trapping, which consists in tight DNA-protein complexes. Here we demonstrate that the cytotoxicity of talazoparib and olaparib results from DNA replication. To elucidate the repair of PARP1-DNA complexes associated with replication in human TK6 and chicken DT40 lymphoblastoid cells, we explored the role of Spartan (SPRTN), a metalloprotease associated with DNA replication, which removes proteins forming DPCs. We find that SPRTN-deficient cells are hypersensitive to talazoparib and olaparib, but not to veliparib, a weak PARP trapper. SPRTN-deficient cells exhibit delayed clearance of trapped PARP1 and increased replication fork stalling upon talazoparib and olaparib treatment. We also show that SPRTN interacts with PARP1 and forms nuclear foci that colocalize with the replicative cell division cycle 45 protein (CDC45) in response to talazoparib. Additionally, SPRTN is deubiquitinated and epistatic with translesion synthesis (TLS) in response to talazoparib. Our results demonstrate that SPRTN is recruited to trapped PARP1 in S-phase to assist in the excision and replication bypass of PARP1-DNA complexes.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Animales , Línea Celular , Pollos , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Humanos , Ftalazinas/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas/toxicidad
4.
Eur J Cancer ; 154: 190-200, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34293664

RESUMEN

BACKGROUND: The impact of maintenance therapy with PARP inhibitors (PARPi) on progression-free survival (PFS) in patients with BRCA mutations and platinum-sensitive recurrent ovarian cancer (PSROC) varies widely. Individual prognostic factors do not reliably distinguish patients who progress early from those who have durable benefit. We developed and validated a prognostic nomogram to predict PFS in these patients. METHODS: The nomogram was developed using data from a training patient cohort with BRCA mutations and high-grade serous PSROC on the placebo arm of two maintenance therapy trials, Study 19 and SOLO2/ENGOT-ov21. We performed multivariable Cox regression analysis based on pre-treatment characteristics to develop a nomogram that predicts PFS. We assessed the discrimination and validation of the nomogram in independent validation patient cohorts treated with maintenance olaparib. RESULTS: The nomogram includes four PFS predictors: CA-125 at randomisation, platinum-free interval, presence of measurable disease and number of prior lines of platinum therapy. In the training (placebo) cohort (internal validation C-index 0.64), median PFS in the model-predicted good, intermediate and poor-risk groups was: 7.7 (95% CI 5.3-11.3), 5.4 (4.8-5.8) and 2.9 (2.8-4.4) months, respectively. In the validation (olaparib) cohort (C-index 0.71), median PFS in the model-predicted good, intermediate and poor-risk groups was: not reached, 16.6 (13.1-22.4) and 8.3 (7.1-10.8) months, respectively. The nomogram showed good calibration in the validation cohort (calibration plot). CONCLUSIONS: This nomogram can be used to predict PFS and counsel patients with BRCA mutations and PSROC prior to maintenance olaparib and for stratification of patients in trials of maintenance therapies.


Asunto(s)
Genes BRCA1 , Genes BRCA2 , Mutación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Nomogramas , Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/toxicidad , Piperazinas/toxicidad , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Pronóstico , Supervivencia sin Progresión
5.
Nucleic Acids Res ; 49(W1): W5-W14, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-33893803

RESUMEN

Because undesirable pharmacokinetics and toxicity of candidate compounds are the main reasons for the failure of drug development, it has been widely recognized that absorption, distribution, metabolism, excretion and toxicity (ADMET) should be evaluated as early as possible. In silico ADMET evaluation models have been developed as an additional tool to assist medicinal chemists in the design and optimization of leads. Here, we announced the release of ADMETlab 2.0, a completely redesigned version of the widely used AMDETlab web server for the predictions of pharmacokinetics and toxicity properties of chemicals, of which the supported ADMET-related endpoints are approximately twice the number of the endpoints in the previous version, including 17 physicochemical properties, 13 medicinal chemistry properties, 23 ADME properties, 27 toxicity endpoints and 8 toxicophore rules (751 substructures). A multi-task graph attention framework was employed to develop the robust and accurate models in ADMETlab 2.0. The batch computation module was provided in response to numerous requests from users, and the representation of the results was further optimized. The ADMETlab 2.0 server is freely available, without registration, at https://admetmesh.scbdd.com/.


Asunto(s)
Farmacocinética , Programas Informáticos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Internet , Preparaciones Farmacéuticas/química , Ftalazinas/química , Ftalazinas/farmacocinética , Ftalazinas/toxicidad , Piperazinas/química , Piperazinas/farmacocinética , Piperazinas/toxicidad
6.
Mol Neurobiol ; 58(8): 3641-3652, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33788167

RESUMEN

Repurposing PARP-1 inhibitors (PARPi) for non-oncological applications offers an attractive therapeutic strategy for pathological conditions characterized by PARP-1 hyperactivity. In the context of Parkinson's disease (PD), PARP-1 hyperactivity has been linked to neuronal death and disease progression. From a therapy perspective, the evaluation of PARPi as neuroprotective agents may offer a new therapeutic alternative for neurodegenerative disorders. An ideal PARPi needs to inhibit PARP-1 hyperactivity while also limiting downstream DNA damage and cellular toxicity-an effect that is attractive in cancer but far from ideal in neurological disease applications. Consequently, in this study, we set out to evaluate the neuroprotective properties of a previously reported low-toxicity PARPi (10e) using in vitro neuronal models of PD. 10e is a structural analogue of FDA-approved PARPi olaparib, with high PARP-1 affinity and selectivity. Our studies revealed that 10e protects neuronal cells from oxidative stress and DNA damage. In addition, 10e exhibits neuroprotective properties against α-synuclein pre-formed fibrils (αSyn PFF) mediated effects, including reduction in the levels of phosphorylated αSyn and protection against abnormal changes in NAD+ levels. Our in vitro studies with 10e provide support for repurposing high-affinity and low-toxicity PARPi for neurological applications and lay the groundwork for long-term therapeutic studies in animal models of PD.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/prevención & control , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Técnicas de Inactivación de Genes/métodos , Humanos , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/toxicidad , Enfermedad de Parkinson/metabolismo , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Ftalazinas/toxicidad , Piperazinas/farmacología , Piperazinas/uso terapéutico , Piperazinas/toxicidad , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/toxicidad
7.
Gan To Kagaku Ryoho ; 47(7): 1089-1092, 2020 Jul.
Artículo en Japonés | MEDLINE | ID: mdl-32668858

RESUMEN

A 43 -year-old woman presented to the hospital with a right breast tumor. She had been treated for human immunodeficiency virus(HIV)infection for 5 years. After being diagnosed with right breast cancer, she underwent total mastectomy and sentinel lymph node biopsy, which indicated T2N1M0 triple-negative breast cancer. She received doxorubicin and cyclophosphamide( AC)followed by docetaxel(AC-T)as postoperative adjuvant chemotherapy. However, 14 months after the adjuvant chemotherapy finished, distant metastasis occurred in the brain, lung, and mediastinum lymph nodes. Treatment for relapse was initiated, with whole brain radiotherapy followed by paclitaxel plus bevacizumab combination therapy(PB); however, new metastatic lesions were found in the bone, liver, and mediastinum lymph node after 2 courses of PB. Given the risk of hereditary breast and ovarian cancer syndrome, a BRCAgene test was performed when the patient received radiotherapy for left recurrent laryngeal nerve paralysis caused by mediastinal lymph nodes; this showed a result positive for a deleterious mutation in BRCA1. Thus, treatment with olaparib, a poly(ADP-ribose)polymerase(PARP)inhibitor, was started. Metastatic lesions, including barky growth, in the liver metastasis were well controlled, as confirmed by CT imaging 4 months after the start of olaparib.


Asunto(s)
Neoplasias de la Mama , Infecciones por VIH , Ftalazinas/toxicidad , Piperazinas/toxicidad , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Infecciones por VIH/complicaciones , Humanos , Mastectomía , Recurrencia Local de Neoplasia
8.
Mar Drugs ; 18(3)2020 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-32244866

RESUMEN

The chemical examination of the marine soft coral Lemnalia sp., collected at the Xisha islands in the South China Sea, resulted in the isolation of four new nardosinane-type sesquiterpenoids, namely clavukoellians G-J (1-4), and one new aristolane sesquiterpene, namely clavukoellian K (5), together with five known compounds, 6-10. The structure elucidation of the isolated natural products was based on various spectroscopic techniques including HRESIMS and NMR, while their absolute configurations were resolved on the basis of comparisons of the ECD spectra with the calculated ECD data. The isolated new compounds 1-5 were evaluated for their anti- and pro- angiogenesis activities in a transgenic fluorescent zebrafish (Tg(vegfr2:GFP)) model. Quantitative analysis revealed that compound 5 displayed pro-angiogenesis activity in a PTK787-induced vascular injury zebrafish model at 2.5 µM. Data showed that compound 5 significantly promoted the angiogenesis in a dose-dependent manner.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Antozoos/química , Productos Biológicos/farmacología , Vasos Sanguíneos/efectos de los fármacos , Sesquiterpenos/farmacología , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/aislamiento & purificación , Inductores de la Angiogénesis/uso terapéutico , Animales , Animales Modificados Genéticamente , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Productos Biológicos/uso terapéutico , China , Espectroscopía de Resonancia Magnética , Modelos Animales , Estructura Molecular , Neovascularización Fisiológica/efectos de los fármacos , Ftalazinas/toxicidad , Piridinas/toxicidad , Sesquiterpenos/química , Sesquiterpenos/aislamiento & purificación , Sesquiterpenos/uso terapéutico , Pez Cebra
9.
Drug Des Devel Ther ; 14: 783-793, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32158196

RESUMEN

BACKGROUND: Talazoparib (BMN673) is a new poly(ADP-ribose) polymerase inhibitor that has been FDA approved for patients suffering from metastatic breast cancer with germline BRCA mutations. METHOD AND RESULTS: In the current study, an accurate and efficient liquid chromatography-tandem mass spectrometry (LC-MS/MS) analytical methodology was developed for TZB estimation in addition to its metabolic stability assessment. TZB and lapatinib (LAP) (which is chosen as an internal standard; IS) were separated using reversed phase elution system (Hypersil C18 column) with an isocratic mobile phase. The linearity range of the established method was 5-500 ng/mL (r2 ≥ 0.999) in the human liver microsomes (HLMs) matrix. Different parameters were calculated to confirm the method sensitivity (limit of quantification was 2.0 ng/mL), and reproducibility (intra- and inter-day precision and accuracy were below 3.1%) of our methodology. For evaluation of TZB metabolic stability in HLM matrix, intrinsic clearance (9.59 µL/min/mg) and in vitro half-life (72.7 mins) were calculated. TZB treatment discontinuations were reported due to adverse events and dose accumulation, so in silico metabolic vulnerability (experimental and in silico) and toxicity assessment (in silico) of TZB were performed utilizing P450 Metabolism and DEREK modules of StarDrop software. CONCLUSION: TZB is slowly metabolized by the liver. TZB was reported to be minimally metabolized by the liver that approved our outcomes. We do recommend that plasma levels be monitored in cases when talazoparib is used for a long period of time, since it is possible for TZB to bioaccumulate after multiple doses to toxic levels. According to our knowledge, the current method is considered the first LC-MS/MS methodology for evaluating TZB metabolic stability. Further drug discovery studies can be done depending on this concept allowing the designing of new series of compounds with more safety profile through reducing side effects and improving metabolic behavior.


Asunto(s)
Simulación por Computador , Ftalazinas/metabolismo , Ftalazinas/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/toxicidad , Calibración , Cromatografía Liquida , Estabilidad de Medicamentos , Humanos , Lapatinib/efectos adversos , Lapatinib/química , Lapatinib/metabolismo , Lapatinib/toxicidad , Microsomas Hepáticos/química , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Estructura Molecular , Ftalazinas/efectos adversos , Ftalazinas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/efectos adversos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Programas Informáticos , Espectrometría de Masas en Tándem
10.
Anticancer Res ; 40(2): 597-608, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32014900

RESUMEN

BACKGROUND/AIM: We performed a systematic review and meta-analysis to investigate the safety of maintenance with olaparib after platinum-based chemotherapy in cancer patients. MATERIALS AND METHODS: Eligible studies included randomized controlled trials (RCTs) regarding the clinical role of olaparib maintenance therapy versus placebo in BRCA-mutated, advanced cancers. Safety profile from each selected study was investigated for all-grade and G3-G4 haematological and non-haematological adverse drug events (ADEs). RESULTS: Four RTCs that involved 1099 patients were included in the analysis. Overall incidences of all-grade and G3-4 ADEs in olaparib group were 97.6% and 41%, respectively. Patients treated with maintenance olaparib showed higher risk of all-grade and G3-G4 anaemia, all-grade neutropenia and thrombocytopenia. Moreover, all-grade and G3-G4 fatigue, all-grade vomiting, diarrhoea, nausea and decreased appetite were more common in the olaparib group compared to placebo. CONCLUSION: Despite an increased risk and incidence of several haematological and non-haematological toxicities, olaparib is a relatively safe agent for the treatment of advanced solid tumors. Prompt identification of ADEs is mandatory to avoid therapy discontinuation and optimize treatment.


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias/tratamiento farmacológico , Ftalazinas/toxicidad , Piperazinas/toxicidad , Placebos/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas/toxicidad , Humanos
11.
Cell Cycle ; 18(15): 1770-1783, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31238782

RESUMEN

Although improvements in radiation therapy were made over the years, radioresistance is still a major challenge. Cancer cells are often deficient for DNA repair response, a feature that is currently exploited as a new anti-cancer strategy. In this context, combination of inhibitors targeting complementary pathways is of interest to sensitize cells to radiation. In this work, we used PARP (Olaparib) and RAD51 (B02) inhibitors to radiosensitize cancer cells to proton and X-ray radiation. More particularly, Olaparib and B02 were used at concentration leading to limited cytotoxic (alone or in combination) but increasing cell death when the cells were irradiated. We showed that, although at limited concentration, Olaparib and B02 were able to radiosensitize different cancer cell lines, i.e. lung and pancreatic cancer cells. Antagonistic, additive or synergistic effects were observed and correlated to cell proliferation rate. The inhibitors enhanced persistent DNA damage, delayed apoptosis, prolonged cell cycle arrest and senescence upon irradiation. These results demonstrated that radiation-induced synthetic lethality might widen the therapeutic window, hence extending the use of PARP inhibitors to patients without BRCAness.


Asunto(s)
Neoplasias/radioterapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Recombinasa Rad51/antagonistas & inhibidores , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Mutaciones Letales Sintéticas , Células A549 , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Compuestos de Boro/uso terapéutico , Compuestos de Boro/toxicidad , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Humanos , Ftalazinas/uso terapéutico , Ftalazinas/toxicidad , Piperazinas/uso terapéutico , Piperazinas/toxicidad , Poli(ADP-Ribosa) Polimerasas/metabolismo , Protones , Radiación Ionizante , Factores de Tiempo
12.
Arch Pharm (Weinheim) ; 352(6): e1800299, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31012160

RESUMEN

To identify new agents for the treatment of American cutaneous leishmaniasis, a series of eight 1,4-bis(substituted benzalhydrazino)phthalazines was evaluated against Leishmania braziliensis and Leishmania mexicana parasites. These compounds represent a disubstituted version of the 1-chloro-4-(monoaryl/heteroarylhydranizyl)phthalazine that exhibited a significant response against L. braziliensis according to our previous findings. Two disubstituted phthalazines 3b and 3f were identified as potential antileishmanial agents against L. braziliensis parasites, exhibiting a submicromolar IC50 response of 2.37 and 7.90 µM on the promastigote form, and of 1.82 and 4.56 µM against intracellular amastigotes, respectively. In particular, compound 3b showed interesting responses against amastigote isolates from reference, glucantime-resistant and clinical human strains, which were by far superior to the biological response found for the glucantime drug. With regard to the toxicity results, both 3b and 3f exhibited moderate LD50 values against murine macrophages (BMDM), with good selectivity indexes on promastigotes and intracellular amastigotes of L. braziliensis. A comparison of biological response was established between the monosubstituted and disubstituted versions of these benzalhydrazino-phthalazines. Easy synthetic procedure and significant response against amastigote strains including against resistant lines made compound 3b a potential candidate for further pharmacokinetic and in vivo experiments as antileishmanial agent, and as a platform for further structural optimization. Mechanism-of-action studies and molecular docking simulations discarded to inhibition of superoxide dismutase as possible mode of action.


Asunto(s)
Antiprotozoarios/farmacología , Leishmania braziliensis/efectos de los fármacos , Leishmania mexicana/efectos de los fármacos , Leishmania/efectos de los fármacos , Ftalazinas/farmacología , Animales , Antiprotozoarios/química , Antiprotozoarios/toxicidad , Células Cultivadas , Leishmania braziliensis/crecimiento & desarrollo , Leishmania braziliensis/metabolismo , Leishmania mexicana/crecimiento & desarrollo , Leishmania mexicana/metabolismo , Dosificación Letal Mediana , Macrófagos/efectos de los fármacos , Macrófagos/parasitología , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Ftalazinas/química , Ftalazinas/toxicidad , Relación Estructura-Actividad , Superóxido Dismutasa/metabolismo
13.
Mol Divers ; 23(2): 283-298, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30168051

RESUMEN

In view of the anticonvulsant activity reported for phthalazine derivatives as non-competitive AMPA receptor antagonists, a new series of phthalazine-1,4-diones (2-12) were designed and synthesized. The neurotoxicity was assessed using rotarod test. The molecular docking was performed for the synthesized compounds to assess their binding affinities toward AMPA receptor as non-competitive antagonists. The molecular modeling data were strongly interrelated to biological screening data. Compounds 8, 7b, 7a, 10 and 3a exhibited the highest binding affinities as non-competitive AMPA receptor antagonists and also showed the highest relative potencies of 1.78, 1.66, 1.60, 1.59 and 1.29, respectively, as anticonvulsants in comparison with diazepam. The most active compounds 8, 7b, 7a, 10 and 3a were further tested against maximal electroshock seizure (MES). Compounds 8 and 7b and 3a showed 100% protection at a dose level of 125 µgm/kg, while compounds 7a and 10 exhibited 83.33% protection at the same dose level. These agents exerted low neurotoxicity and high safety margin in comparison with valproate as a reference drug. Most of our designed compounds exhibited good ADMET profile.


Asunto(s)
Anticonvulsivantes , Ftalazinas , Receptores AMPA/antagonistas & inhibidores , Animales , Anticonvulsivantes/química , Anticonvulsivantes/farmacocinética , Anticonvulsivantes/uso terapéutico , Anticonvulsivantes/toxicidad , Diseño de Fármacos , Electrochoque , Ratones , Modelos Biológicos , Simulación del Acoplamiento Molecular , Ftalazinas/química , Ftalazinas/farmacocinética , Ftalazinas/uso terapéutico , Ftalazinas/toxicidad , Convulsiones/tratamiento farmacológico , Relación Estructura-Actividad
14.
PLoS One ; 12(6): e0179278, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28628639

RESUMEN

Flap endonuclease 1 (FEN1) is a structure selective endonuclease required for proficient DNA replication and the repair of DNA damage. Cellularly active inhibitors of this enzyme have previously been shown to induce a DNA damage response and, ultimately, cell death. High-throughput screens of human cancer cell-lines identify colorectal and gastric cell-lines with microsatellite instability (MSI) as enriched for cellular sensitivity to N-hydroxyurea series inhibitors of FEN1, but not the PARP inhibitor olaparib or other inhibitors of the DNA damage response. This sensitivity is due to a synthetic lethal interaction between FEN1 and MRE11A, which is often mutated in MSI cancers through instabilities at a poly(T) microsatellite repeat. Disruption of ATM is similarly synthetic lethal with FEN1 inhibition, suggesting that disruption of FEN1 function leads to the accumulation of DNA double-strand breaks. These are likely a result of the accumulation of aberrant replication forks, that accumulate as a consequence of a failure in Okazaki fragment maturation, as inhibition of FEN1 is toxic in cells disrupted for the Fanconi anemia pathway and post-replication repair. Furthermore, RAD51 foci accumulate as a consequence of FEN1 inhibition and the toxicity of FEN1 inhibitors increases in cells disrupted for the homologous recombination pathway, suggesting a role for homologous recombination in the resolution of damage induced by FEN1 inhibition. Finally, FEN1 appears to be required for the repair of damage induced by olaparib and cisplatin within the Fanconi anemia pathway, and may play a role in the repair of damage associated with its own disruption.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Endonucleasas de ADN Solapado/metabolismo , Hidroxiurea/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/toxicidad , ADN/efectos de los fármacos , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Endonucleasas de ADN Solapado/antagonistas & inhibidores , Endonucleasas de ADN Solapado/genética , Humanos , Hidroxiurea/química , Proteína Homóloga de MRE11 , Inestabilidad de Microsatélites/efectos de los fármacos , Mutación , Ftalazinas/toxicidad , Piperazinas/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas/toxicidad , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/genética
15.
Mol Cancer Res ; 14(12): 1195-1203, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27604276

RESUMEN

Many drugs targeting the DNA damage response are being developed as anticancer therapies, either as single agents or in combination with ionizing radiation (IR) or other cytotoxic agents. Numerous clinical trials in this area are either in progress or planned. However, concerns remain about the potential of such treatments to increase toxicity to normal tissues. In order to address this issue, a novel reporter mouse line was created through the simultaneous incorporation of multiple reporters, ß-galactosidase, and firefly luciferase, into the DNA damage-inducible p21 (CDKN1A) locus. The data demonstrate that in situ ß-galactosidase staining facilitates high fidelity mapping of p21 expression across multiple organs and tissues at single-cell resolution, whereas the luciferase reporter permits noninvasive bioluminescent imaging of p21 expression. This model was used to determine the capacity of a number of DNA-damaging agents, including IR, cisplatin, and etoposide to induce p21 expression in normal tissues. In addition, the PARP inhibitor olaparib was examined alone or in combination with IR as well as cisplatin. A single exposure to olaparib alone caused DNA damage to cells in the mucosal layer lining mouse large intestine. It also exacerbated DNA damage induced in this organ and the kidney by coadministered IR. These studies suggest that olaparib has carcinogenic potential and illustrate the power of this new model to evaluate the safety of new therapeutic regimens involving combination therapies. IMPLICATIONS: Olaparib causes DNA damage to normal tissues and might be a carcinogen. Mol Cancer Res; 14(12); 1195-203. ©2016 AACR.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Mucosa Intestinal/efectos de los fármacos , Luciferasas de Luciérnaga/metabolismo , Ftalazinas/toxicidad , Piperazinas/toxicidad , beta-Galactosidasa/metabolismo , Animales , Línea Celular Tumoral , Cisplatino/toxicidad , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Luciferasas de Luciérnaga/genética , Ratones , Ratones Transgénicos , Radiación Ionizante , Análisis de la Célula Individual , beta-Galactosidasa/genética
16.
Cancer Res ; 76(20): 6084-6094, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27550455

RESUMEN

The PARP inhibitor AZD2461 was developed as a next-generation agent following olaparib, the first PARP inhibitor approved for cancer therapy. In BRCA1-deficient mouse models, olaparib resistance predominantly involves overexpression of P-glycoprotein, so AZD2461 was developed as a poor substrate for drug transporters. Here we demonstrate the efficacy of this compound against olaparib-resistant tumors that overexpress P-glycoprotein. In addition, AZD2461 was better tolerated in combination with chemotherapy than olaparib in mice, which suggests that AZD2461 could have significant advantages over olaparib in the clinic. However, this superior toxicity profile did not extend to rats. Investigations of this difference revealed a differential PARP3 inhibitory activity for each compound and a higher level of PARP3 expression in bone marrow cells from mice as compared with rats and humans. Our findings have implications for the use of mouse models to assess bone marrow toxicity for DNA-damaging agents and inhibitors of the DNA damage response. Finally, structural modeling of the PARP3-active site with different PARP inhibitors also highlights the potential to develop compounds with different PARP family member specificity profiles for optimal antitumor activity and tolerability. Cancer Res; 76(20); 6084-94. ©2016 AACR.


Asunto(s)
Neoplasias Experimentales/tratamiento farmacológico , Ftalazinas/farmacología , Piperidinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/fisiología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Animales , Médula Ósea/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Reparación del ADN , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Descubrimiento de Drogas , Genes BRCA1 , Humanos , Ratones , Ftalazinas/administración & dosificación , Ftalazinas/toxicidad , Piperazinas/administración & dosificación , Piperidinas/toxicidad , Poli(ADP-Ribosa) Polimerasas/química , Ratas , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
17.
ChemMedChem ; 11(10): 1078-89, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27097919

RESUMEN

Tuberculosis (TB) remains one of the major causes of death worldwide, in particular because of the emergence of multidrug-resistant TB. Herein we explored the potential of an alternative class of molecules as anti-TB agents. Thus, a series of novel 3-substituted triazolophthalazines was quickly and easily prepared from commercial hydralazine hydrochloride as starting material and were further evaluated for their antimycobacterial activities and cytotoxicities. Four of the synthesized compounds were found to effectively inhibit the Mycobacterium tuberculosis (M.tb) H37 Rv strain with minimum inhibitory concentration (MIC) values <10 µg mL(-1) , whereas no compounds displayed cytotoxicity against HCT116 human cell lines (IC50 >100 µm). More remarkably, the most potent compounds proved to be active to a similar extent against various multidrug-resistant M.tb strains, thus uncovering a mode of action distinct from that of standard antitubercular agents. Overall, their ease of preparation, combined with their attractive antimycobacterial activities, make such triazolophthalazine-based derivatives promising leads for further development.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Ftalazinas/farmacología , Antituberculosos/química , Antituberculosos/toxicidad , Supervivencia Celular/efectos de los fármacos , Cinamatos/química , Farmacorresistencia Bacteriana/efectos de los fármacos , Células HCT116 , Humanos , Pruebas de Sensibilidad Microbiana , Ftalazinas/química , Ftalazinas/toxicidad , Tuberculosis
18.
Cell Physiol Biochem ; 37(4): 1537-46, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26512879

RESUMEN

BACKGROUND/AIMS: The aldose reductase inhibitor zopolrestat has been shown to either decrease or increase apoptosis, the suicidal death of nucleated cells. Erythrocytes may similarly enter suicidal death or eryptosis, which is characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Triggers of eryptosis include oxidative stress, Ca2+ entry with increase of cytosolic Ca2+ activity ([Ca2+]i), and ceramide formation. The present study explored, whether and how zopolrestat induces eryptosis. METHODS: Phosphatidylserine exposure at the cell surface was estimated from annexin V binding, cell volume from forward scatter, oxidative stress from DCFDA dependent fluorescence, [Ca2+]i from Fluo3-fluorescence, and ceramide abundance utilizing specific antibodies. RESULTS: A 48 hours exposure of human erythrocytes to zopolrestat (≥ 150 µg/ml) significantly increased the percentage of annexin-V-binding cells, significantly decreased forward scatter (≥ 125 µg/ml), significantly increased Fluo3-fluorescence (200 µg/ml), significantly increased ceramide abundance (150 µg/ml), but did not significantly modify DCFDA fluorescence. The effect of zopolrestat on annexin-V-binding was significantly blunted, but not abolished by removal of extracellular Ca2+. CONCLUSIONS: Exposure of human erythrocytes to zopolrestat triggers cell shrinkage and cell membrane scrambling, an effect in part due to Ca2+ entry and ceramide.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzotiazoles/toxicidad , Inhibidores Enzimáticos/toxicidad , Ftalazinas/toxicidad , Compuestos de Anilina/química , Calcio/metabolismo , Tamaño de la Célula/efectos de los fármacos , Ceramidas/análisis , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Especies Reactivas de Oxígeno/metabolismo , Xantenos/química
19.
Anticancer Res ; 34(10): 5487-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25275045

RESUMEN

BACKGROUND/AIM: Gemtuzumab ozogamicin (GO) consists of the cluster of differentiation 33 (CD33) antibody linked to calicheamicin. The binding of GO to the CD33 antigen on leukemic cells results in internalization and subsequent release of calicheamicin, thereby inducing DNA strand breaks. We hypothesized that the poly (ADP-ribose) polymerase inhibitor olaparib might inhibit DNA repair initiated by GO-induced DNA strand breaks, thereby increasing cytotoxicity. MATERIALS AND METHODS: The human myeloid leukemia cell line HL-60 and a GO-resistant variant (HL/GO20) were used. RESULTS: The 50% growth-inhibitory concentrations (IC50) were 24 ng/ml for HL-60 cells and 550 ng/ml for GO-resistant variant HL/GO20 cells. HL/GO20 cells were also refractory to GO-induced apoptosis. CD33 positivity was reduced in HL/GO20 cells. Olaparib-alone did not inhibit the cell growth and did not induce apoptosis in either HL-60 cells or HL/GO20 cells at concentrations of up to 10 µM. When cells were treated with different concentrations of GO in the presence of 10 µM olaparib, the IC50 of GO for HL-60 cells was 13 ng/ml. The combination index was 0.86, indicating synergistic cytotoxicity of GO and olaparib in combination. Such a combination was ineffective for HL/GO20 cells. CONCLUSION: GO and olaparib exerted synergistic cytotoxicity in CD33-positive myeloid leukemia cells in vitro.


Asunto(s)
Aminoglicósidos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Leucemia Mieloide/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Aminoglicósidos/toxicidad , Anticuerpos Monoclonales Humanizados/toxicidad , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Gemtuzumab , Células HL-60 , Humanos , Ftalazinas/toxicidad , Piperazinas/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas
20.
Blood ; 121(1): 54-63, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23093618

RESUMEN

SLX4, the newly identified Fanconi anemia protein, FANCP, is implicated in repairing DNA damage induced by DNA interstrand cross-linking (ICL) agents, topoisomerase I (TOP1) inhibitors, and in Holliday junction resolution. It interacts with and enhances the activity of XPF-ERCC1, MUS81-EME1, and SLX1 nucleases, but the requirement for the specific nucleases in SLX4 function is unclear. Here, by complementing a null FA-P Fanconi anemia cell line with SLX4 mutants that specifically lack the interaction with each of the nucleases, we show that the SLX4-dependent XPF-ERCC1 activity is essential for ICL repair but is dispensable for repairing TOP1 inhibitor-induced DNA lesions. Conversely, MUS81-SLX4 interaction is critical for resistance to TOP1 inhibitors but is less important for ICL repair. Mutation of SLX4 that abrogates interaction with SLX1 results in partial resistance to both cross-linking agents and TOP1 inhibitors. These results demonstrate that SLX4 modulates multiple DNA repair pathways by regulating appropriate nucleases.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN/fisiología , Anemia de Fanconi/genética , Recombinasas/fisiología , Camptotecina/toxicidad , Línea Celular , Reactivos de Enlaces Cruzados/toxicidad , ADN/efectos de los fármacos , ADN/metabolismo , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas , Endonucleasas/metabolismo , Anemia de Fanconi/enzimología , Anemia de Fanconi/patología , Humanos , Mitomicina/toxicidad , Ftalazinas/toxicidad , Piperazinas/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Recombinasas/deficiencia , Recombinasas/genética , Inhibidores de Topoisomerasa I/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...