Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Yao Xue Xue Bao ; 48(7): 1053-70, 2013 Jul.
Artigo em Zh | MEDLINE | ID: mdl-24133971

RESUMO

Chemotherapy remains one of the major tools, along with surgery, radiotherapy, and more recently targeted therapy, in the war against cancer. There have appeared a plethora of highly potent cytotoxic drugs but the poor discriminability between cancerous and healthy cells of these agents limits their broader application in clinical settings. Therapeutic antibodies have emerged as an important class of biological anticancer agents, thanks to their ability in specific binding to tumor-associated antigens. While this important class of biologics can be used as single agents for the treatment of cancer through antibody-dependent cell cytotoxicity (ADCC), their therapeutical efficacy is often limited. Antitumor antibody drug conjugates (ADCs) combine the target-specificity of monoclonal antibody (mAb) and the highly active cell-killing drugs, taking advantages of the best characteristics out of both components. Thus, insufficiency of most naked mAbs in cancer therapy has been circumvented by arming the immunoglobulin with cytotoxic drugs. Here mAbs are used as vehicles to transport potent payloads to tumor cells. ADCs contain three main components: antibody, linker and cytotoxics (also frequently referred as payload). Antibodies can recognize and specifically bind to the tumor-specific antigens, leading to an antibody-assisted internalization, and payload release. While ADC has demonstrated tremendous success, a number of practical challenges limit the broader applications of this new class of anticancer therapy, including inefficient cellular uptake, low cytotoxicity, and off-target effects. This review article aims to cover recent advances in optimizing linkers with increased stability in circulation while allowing efficient payload release within tumor cells. We also attempt to provide some practical strategies in resolving the current challenges in this attractive research area, particularly to those new to the field.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Citotoxinas/uso terapêutico , Desenho de Fármacos , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Aminobenzoatos/farmacologia , Aminobenzoatos/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Citotoxinas/farmacologia , Humanos , Imunoconjugados/química , Imunoconjugados/farmacologia , Maitansina/farmacologia , Maitansina/uso terapêutico , Neoplasias/patologia , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico
2.
Bioorg Med Chem ; 20(15): 4635-45, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22766219

RESUMO

PARP-1, the most abundant member of the PARP superfamily of nuclear enzymes, has emerged as a promising molecular target in the past decade particularly for the treatment of cancer. A number of PARP-1 inhibitors, including veliparab discovered at Abbott, have advanced into different stages of clinical trials. Herein we describe the development of a new tetrahydropyridopyridazinone series of PARP-1 inhibitors. Many compounds in this class, such as 20w, displayed excellent potency against the PARP-1 enzyme with a K(i) value of <1nM and an EC(50) value of 1nM in a C41 whole cell assay. The presence of the NH in the tetrahydropyridyl ring of the tetrahydropyridopyridazinone scaffold improved the pharmacokinetic properties over similar carbon based analogs. Compounds 8c and 20u are orally available, and have demonstrated significant efficacy in a B16 murine xenograft model, potentiating the efficacy of temozolomide (TMZ).


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases , Piridazinas/farmacologia , Piridinas/farmacologia , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/síntese química , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/síntese química , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Estrutura Molecular , Neoplasias Experimentais/enzimologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Piridazinas/síntese química , Piridazinas/química , Piridinas/síntese química , Piridinas/química , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
3.
ACS Med Chem Lett ; 12(7): 1108-1115, 2021 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-34267880

RESUMO

Cyclin-dependent kinase 9 (CDK9) is a serine/threonine kinase involved in the regulation of transcription elongation. An inhibition of CDK9 downregulates a number of short-lived proteins responsible for tumor maintenance and survival, including the antiapoptotic BCL-2 family member MCL-1. As pan-CDK inhibitors under development have faced dosing and toxicity challenges in the clinical setting, we generated selective CDK9 inhibitors that could be amenable to an oral administration. Here, we report the lead optimization of a series of azaindole-based inhibitors. To overcome early challenges with promiscuity and cardiovascular toxicity, carboxylates were introduced into the pharmacophore en route to compounds such as 14 and 16. These CDK9 inhibitors demonstrated a reduced toxicity, adequate pharmacokinetic properties, and a robust in vivo efficacy in mice upon oral dosing.

4.
Bioorg Med Chem Lett ; 20(3): 1023-6, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20045315

RESUMO

Through conformational restriction of a benzamide by formation of a seven-membered hydrogen-bond with an oxindole carbonyl group, a series of PARP inhibitors was designed for appropriate orientation for binding to the PARP surface. This series of compounds with a 3-oxoisoindoline-4-carboxamide core structure, displayed modest to good activity against PARP-1 in both intrinsic and cellular assays. SAR studies at the lactam nitrogen of the pharmacophore have suggested that a secondary or tertiary amine is important for cellular potency. An X-ray structure of compound 1e bound to the protein confirmed the formation of a seven-membered intramolecular hydrogen bond. Though revealed previously in peptides, this type of seven-membered intramolecular hydrogen bond is rarely observed in small molecules. Largely due to the formation of the intramolecular hydrogen bond, the 3-oxoisoindoline-4-carboxamide core structure appears to be planar in the X-ray structure. An additional hydrogen bond interaction of the piperidine nitrogen to Gly-888 also contributes to the binding affinity of 1e to PARP-1.


Assuntos
Amidas/química , Antineoplásicos/química , Descoberta de Drogas/métodos , Isoindóis/química , Neoplasias/enzimologia , Inibidores de Poli(ADP-Ribose) Polimerases , Amidas/metabolismo , Amidas/uso terapêutico , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Cristalografia por Raios X , Isoindóis/metabolismo , Isoindóis/uso terapêutico , Neoplasias/tratamento farmacológico , Poli(ADP-Ribose) Polimerases/metabolismo , Relação Estrutura-Atividade
5.
Mol Cancer Res ; 6(10): 1621-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18922977

RESUMO

Poly(ADP-ribose) polymerase (PARP) senses DNA breaks and facilitates DNA repair via the polyADP-ribosylation of various DNA binding and repair proteins. We explored the mechanism of potentiation of temozolomide cytotoxicity by the PARP inhibitor ABT-888. We showed that cells treated with temozolomide need to be exposed to ABT-888 for at least 17 to 24 hours to achieve maximal cytotoxicity. The extent of cytotoxicity correlates with the level of double-stranded DNA breaks as indicated by gammaH2AX levels. In synchronized cells, damaging DNA with temozolomide in the presence of ABT-888 during the S phase generated high levels of double-stranded breaks, presumably because the single-stranded DNA breaks resulting from the cleavage of the methylated nucleotides were converted into double-stranded breaks through DNA replication. As a result, treatment of temozolomide and ABT-888 during the S phase leads to higher levels of cytotoxicity. ABT-888 inhibits poly(ADP-ribose) formation in vivo and enhances tumor growth inhibition by temozolomide in multiple models. ABT-888 is well tolerated in animal models. ABT-888 is currently in clinical trials in combination with temozolomide.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , Dacarbazina/análogos & derivados , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Dacarbazina/farmacologia , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Humanos , Camundongos , Ratos , Temozolomida
6.
Bioorg Med Chem Lett ; 18(14): 3955-8, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18586490

RESUMO

Poly(ADP-ribose) polymerases (PARPs) play significant roles in various cellular functions including DNA repair and control of RNA transcription. PARP inhibitors have been demonstrated to potentiate the effect of cytotoxic agents or radiation in a number of animal tumor models. Utilizing a benzimidazole carboxamide scaffold in which the amide forms a key intramolecular hydrogen bond for optimal interaction with the enzyme, we have identified a novel series of PARP inhibitors containing a quaternary methylene-amino substituent at the C-2 position of the benzimidazole. Geminal dimethyl analogs at the methylene-amino substituent were typically more potent than mono-methyl derivatives in both intrinsic and cellular assays. Smaller cycloalkanes such as cyclopropyl or cyclobutyl were tolerated at the quaternary carbon while larger rings were detrimental to potency. In vivo efficacy data in a B16F10 murine flank melanoma model in combination with temozolomide (TMZ) are described for two optimized analogs.


Assuntos
Antineoplásicos/síntese química , Química Farmacêutica/instrumentação , Inibidores Enzimáticos/síntese química , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Química Farmacêutica/métodos , DNA/química , Reparo do DNA , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/farmacologia , Cinética , Camundongos , Transplante de Neoplasias , Transcrição Gênica
7.
Anticancer Res ; 28(5A): 2625-35, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19035287

RESUMO

ABT-888 is a potent, orally bioavailable PARP-1/2 inhibitor shown to potentiate DNA damaging agents. The ability to potentiate temozolomide (TMZ) and develop a biological marker for PARP inhibition was evaluated in vivo. Doses/schedules that achieve TMZ potentiation in the B16F10 syngeneic melanoma model were utilized to develop an ELISA to detect a pharmacodynamic marker, ADP ribose polymers (pADPr), after ABT 888 treatment. ABT-888 enhanced TMZ antitumor activity, in a dose-proportional manner with no observed toxicity (44-75% tumor growth inhibition vs. TMZ monotherapy), but did not show single agent activity. Extended ABT-888 dosing schedules showed no advantage compared to simultaneous TMZ administration. Efficacy correlated with plasma/tumor drug concentrations. Intratumor drug levels correlated with a dose-proportional/time-dependent reduction in pADPr. Potentiation of TMZ activity by ABT-888 correlated with drug levels and inhibition of PARP activity in vivo. ABT-888 is in Phase 1 trials using a validated ELISA based on the assay developed here to assess pharmacological effect.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzimidazóis/farmacologia , Dacarbazina/análogos & derivados , Melanoma Experimental/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Benzimidazóis/administração & dosagem , Benzimidazóis/farmacocinética , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Esquema de Medicação , Sinergismo Farmacológico , Melanoma Experimental/enzimologia , Melanoma Experimental/metabolismo , Camundongos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Temozolomida
8.
Bioorg Med Chem ; 16(14): 6965-75, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18541433

RESUMO

We have developed a series of cyclic amine-containing benzimidazole carboxamide poly(ADP-ribose)polymerase (PARP) inhibitors, with good PARP-1 enzyme potency, as well as cellular potency. These efforts led to the identification of a lead preclinical candidate, 10b, 2-(1-propylpiperidin-4-yl)-1H-benzimidazole-4-carboxamide (A-620223). 10b displayed very good potency against both the PARP-1 enzyme with a K(i) of 8nM and in a whole cell assay with an EC(50) of 3nM. 10b is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide (TMZ) and in an MX-1 breast xenograph model in combination with cisplatin.


Assuntos
Benzimidazóis/química , Benzimidazóis/farmacologia , Neoplasias da Mama/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/uso terapêutico , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Relação Estrutura-Atividade , Temozolomida , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Clin Cancer Res ; 13(9): 2728-37, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17473206

RESUMO

PURPOSE: To evaluate the preclinical pharmacokinetics and antitumor efficacy of a novel orally bioavailable poly(ADP-ribose) polymerase (PARP) inhibitor, ABT-888. EXPERIMENTAL DESIGN: In vitro potency was determined in a PARP-1 and PARP-2 enzyme assay. In vivo efficacy was evaluated in syngeneic and xenograft models in combination with temozolomide, platinums, cyclophosphamide, and ionizing radiation. RESULTS: ABT-888 is a potent inhibitor of both PARP-1 and PARP-2 with K(i)s of 5.2 and 2.9 nmol/L, respectively. The compound has good oral bioavailability and crosses the blood-brain barrier. ABT-888 strongly potentiated temozolomide in the B16F10 s.c. murine melanoma model. PARP inhibition dramatically increased the efficacy of temozolomide at ABT-888 doses as low as 3.1 mg/kg/d and a maximal efficacy achieved at 25 mg/kg/d. In the 9L orthotopic rat glioma model, temozolomide alone exhibited minimal efficacy, whereas ABT-888, when combined with temozolomide, significantly slowed tumor progression. In the MX-1 breast xenograft model (BRCA1 deletion and BRCA2 mutation), ABT-888 potentiated cisplatin, carboplatin, and cyclophosphamide, causing regression of established tumors, whereas with comparable doses of cytotoxic agents alone, only modest tumor inhibition was exhibited. Finally, ABT-888 potentiated radiation (2 Gy/d x 10) in an HCT-116 colon carcinoma model. In each model, ABT-888 did not display single-agent activity. CONCLUSIONS: ABT-888 is a potent inhibitor of PARP, has good oral bioavailability, can cross the blood-brain barrier, and potentiates temozolomide, platinums, cyclophosphamide, and radiation in syngeneic and xenograft tumor models. This broad spectrum of chemopotentiation and radiopotentiation makes this compound an attractive candidate for clinical evaluation.


Assuntos
Benzimidazóis/administração & dosagem , Benzimidazóis/farmacocinética , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Neoplasias/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases , Administração Oral , Animais , Antineoplásicos Alquilantes/uso terapêutico , Disponibilidade Biológica , Barreira Hematoencefálica/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Modelos Animais de Doenças , Cães , Sinergismo Farmacológico , Feminino , Haplorrinos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Ratos , Ratos Endogâmicos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Front Neurosci ; 12: 889, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30546294

RESUMO

Obsessive-compulsive disorder (OCD) patients have difficulty in switching between obsessive thought and compulsive behavior, which may be related to the dysfunction of the salience network (SN). However, little is known about the changes in intra- and inter- intrinsic functional connectivity (iFC) of the SN in patients with OCD. In this study, we parceled the SN into 19 subregions and investigated iFC changes for each of these subregions in 40 drug-naïve patients with OCD and 40 healthy controls (HCs) using seed-based functional connectivity resting-state functional magnetic resonance imaging (rs-fMRI). We found that patients with OCD exhibited decreased iFC strength between subregions of the SN, as well as decreased inter-network connectivity between SN and DMN, and ECN. These findings highlight a specific alteration in iFC patterns associated with SN in patients with OCD and provide new insights into the dysfunctional brain organization of the SN in patients with OCD.

11.
J Med Chem ; 50(13): 2990-3003, 2007 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-17523610

RESUMO

Compound 7 was identified as a potent (IC50 = 14 nM), selective, and orally bioavailable (F = 70% in mouse) inhibitor of protein kinase B/Akt. While promising efficacy was observed in vivo, this compound showed effects on depolarization of Purkinje fibers in an in vitro assay and CV hypotension in vivo. Guided by an X-ray structure of 7 bound to protein kinase A, which has 80% homology with Akt in the kinase domain, our efforts have focused on structure-activity relationship (SAR) studies of the phenyl moiety, in an attempt to address the cardiovascular liability and further improve the Akt potency. A novel and efficient synthetic route toward diversely substituted phenyl derivatives of 7 was developed utilizing a copper-mediated aziridine ring-opening reaction as the key step. To improve the selectivity of these Akt inhibitors over other protein kinases, a nitrogen atom was incorporated into selected phenyl analogues of 7 at the C-6 position of the methyl indazole scaffold. These modifications resulted in the discovery of inhibitor 37c with greater potency (IC50 = 0.6 nM vs Akt), selectivity, and improved cardiovascular safety profile. The SARs, pharmacokinetic profile, and CV safety of selected Akt inhibitors will be discussed.


Assuntos
Hipotensão/induzido quimicamente , Indazóis/síntese química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Pirazóis/síntese química , Piridinas/síntese química , Administração Oral , Animais , Disponibilidade Biológica , Cristalografia por Raios X , Cães , Indazóis/efeitos adversos , Indazóis/farmacologia , Camundongos , Modelos Moleculares , Conformação Proteica , Ramos Subendocárdicos/efeitos dos fármacos , Ramos Subendocárdicos/fisiologia , Pirazóis/efeitos adversos , Pirazóis/farmacologia , Piridinas/efeitos adversos , Piridinas/farmacologia , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
12.
Curr Top Med Chem ; 2(9): 939-71, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12171565

RESUMO

Over the past decade, protein kinases have emerged as a group of molecular targets with the potential to be "cancer-specific", allowing the selective targeting of cancer cells versus normal cells. These selective anticancer drugs would eliminate the cytotoxic side effects that are associated with conventional cancer chemotherapy. This article will focus on two emerging and less-explored protein serine/threonine kinase targets: PKB/Akt and checkpoint kinase 1 (Chk1). Protein kinase B/Akts are a group of serine/threonine kinases that are overexpressed in a variety of human tumors. An Akt inhibitor would target the imbalance of pro-versus anti-apoptosis regulation in cancerous as compared to healthy cells. Thus, a greater therapeutic window than conventional cytotoxic chemotherapy is expected. Cell-cycle checkpoints have become attractive targets since some of them, such as the G1/S checkpoint, are defective in most tumor cells. Inhibition of one or more of the remaining checkpoint(s) could make cancerous cells more sensitive than healthy cells toward DNA damaging agents or radiation therapy. Among the checkpoint kinases, Chk1 appears to be an attractive molecular target. Chk1 blocks the activation of the Cdc2-cyclin B kinase complex, and hence entry into mitosis, by disrupting the translocation of the phosphatase Cdc25C from the cyotoplasm to the nucleus. A limited number of small molecule inhibitors in this emerging field and their mode of action will be reviewed.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Ciclo Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Dano ao DNA/efeitos dos fármacos , Inibidores Enzimáticos/química , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Neoplasias/enzimologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Homologia de Sequência de Aminoácidos
13.
Clin Exp Med ; 12(3): 135-43, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21874584

RESUMO

Gaucher disease is an autosomal recessive lysosomal storage disorder resulting in a deficiency of glucocerebrosidase (GC). Imiglucerase, a recombinant form of GC, has been successfully used in the treatment of Gaucher disease and has been shown to be a good potential candidate for gene therapy. However, its low transduction efficiency and short duration of expression have limited it as a gene therapy strategy. VP22, the herpes simplex virus type I tegument protein, is known to facilitate intercellular protein transport, thus making it a promising tool for improving gene transfer efficiency. To investigate whether the fusion of VP22 to GC could improve its therapeutic efficiency for Gaucher disease, the lentiviral vectors pHIV-GC and pHIV-VP(22)-GC were constructed and confirmed by PCR or RT-PCR. After packaging, the vectors were transduced into human Gaucher II fibroblast cells (GII cells). Flow cytometric analysis revealed that the GC expression rates in lenti-VP(22)-GC-transduced GII cells were higher by comparison than those in lenti-GC-transduced GII cells. A Western blot demonstrated higher levels of GC protein expression in lenti-VP(22)-GC-transduced GII cells. In addition, the long-term expression levels and increased GC activities in lenti-VP(22)-GC-transduced GII cells were also observed. These data implicate that VP22-mediated effects may be useful for enhancing the efficacy of this Gaucher disease treatment.


Assuntos
Fibroblastos/enzimologia , Doença de Gaucher/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Glucosilceramidase/metabolismo , Proteínas Estruturais Virais/metabolismo , Ativação Enzimática , Fibroblastos/citologia , Citometria de Fluxo , Imunofluorescência , Doença de Gaucher/genética , Regulação Enzimológica da Expressão Gênica , Glucosilceramidase/genética , Células HEK293 , Herpesvirus Humano 1/genética , Humanos , Lentivirus/genética , Plasmídeos/genética , Plasmídeos/metabolismo , Cultura Primária de Células , Transfecção , Proteínas Estruturais Virais/genética
14.
J Med Chem ; 53(8): 3142-53, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20337371

RESUMO

We have developed a series of phenylpyrrolidine- and phenylpiperidine-substituted benzimidazole carboxamide poly(ADP-ribose) polymerase (PARP) inhibitors with excellent PARP enzyme potency as well as single-digit nanomolar cellular potency. These efforts led to the identification of (S)-2-(2-fluoro-4-(pyrrolidin-2-yl)phenyl)-1H-benzimidazole-4-carboxamide (22b, A-966492). Compound 22b displayed excellent potency against the PARP-1 enzyme with a K(i) of 1 nM and an EC(50) of 1 nM in a whole cell assay. In addition, 22b is orally bioavailable across multiple species, crosses the blood-brain barrier, and appears to distribute into tumor tissue. It also demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide and in an MX-1 breast cancer xenograft model both as a single agent and in combination with carboplatin.


Assuntos
Antineoplásicos/síntese química , Benzimidazóis/síntese química , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína BRCA1/deficiência , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Disponibilidade Biológica , Barreira Hematoencefálica/metabolismo , Carboplatina/administração & dosagem , Linhagem Celular Tumoral , Cristalografia por Raios X , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Melanoma Experimental/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Modelos Moleculares , Transplante de Neoplasias , Estereoisomerismo , Relação Estrutura-Atividade , Temozolomida , Transplante Heterólogo
15.
J Med Chem ; 52(2): 514-23, 2009 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19143569

RESUMO

We have developed a series of cyclic amine-containing benzimidazole carboxamide PARP inhibitors with a methyl-substituted quaternary center at the point of attachment to the benzimidazole ring system. These compounds exhibit excellent PARP enzyme potency as well as single-digit nanomolar cellular potency. These efforts led to the identification of 3a (2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide, ABT-888), currently in human phase I clinical trials. Compound 3a displayed excellent potency against both the PARP-1 and PARP-2 enzymes with a K(i) of 5 nM and in a C41 whole cell assay with an EC(50) of 2 nM. In addition, 3a is aqueous soluble, orally bioavailable across multiple species, and demonstrated good in vivo efficacy in a B16F10 subcutaneous murine melanoma model in combination with temozolomide (TMZ) and in an MX-1 breast cancer xenograft model in combination with either carboplatin or cyclophosphamide.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Melanoma Experimental/patologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Área Sob a Curva , Benzimidazóis/administração & dosagem , Benzimidazóis/farmacocinética , Disponibilidade Biológica , Carboplatina/administração & dosagem , Ciclofosfamida/administração & dosagem , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Cães , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Feminino , Haplorrinos , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos SCID , Ratos , Temozolomida
16.
Mol Cancer Res ; 7(10): 1686-92, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19825992

RESUMO

Many established cancer therapies involve DNA-damaging chemotherapy or radiotherapy. Gain of DNA repair capacity of the tumor represents a common mechanism used by cancer cells to survive DNA-damaging therapy. Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear enzyme that is activated by DNA damage and plays a critical role in base excision repair. Inhibition of PARP represents an attractive approach for the treatment of cancer. Previously, we have described the discovery and characterization of a potent PARP inhibitor, ABT-888. ABT-888 potentiates the activity of DNA-damaging agents such as temozolomide (TMZ) in a variety of preclinical models. We report here the generation of HCT116 cells resistant to treatment with TMZ and ABT-888 (HCT116R cells). HCT116R cells exhibit decreased H2AX phosphorylation in response to treatment with TMZ and ABT-888 relative to parental HCT116 cells. Microarray and Western blot studies indicate that HCT116R cells have decreased PARP-1 and elevated Rad51 expression levels. HCT116R cells are dependent on Rad51 for proliferation and survival, as shown by inhibition of proliferation and induction of apoptosis upon treatment with Rad51 small interfering RNA. In addition, HCT116R cells are more resistant to radiation than the parental HCT116 cells. Our study suggests that cancer cells upregulate the homologous recombination DNA repair pathway to compensate for the loss of base excision repair, which may account for the observed resistance to treatment with TMZ and ABT-888.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzimidazóis/farmacologia , Reparo do DNA/efeitos dos fármacos , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/genética , Recombinação Genética/efeitos dos fármacos , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Reparo do DNA/genética , Dacarbazina/farmacologia , Regulação para Baixo/genética , Histonas/efeitos dos fármacos , Histonas/genética , Histonas/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Interferente Pequeno , Rad51 Recombinase/efeitos dos fármacos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Recombinação Genética/genética , Homologia de Sequência , Temozolomida
17.
Clin Cancer Res ; 15(23): 7277-90, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19934293

RESUMO

PURPOSE: ABT-888, currently in phase 2 trials, is a potent oral poly(ADP-ribose) polymerase inhibitor that enhances the activity of multiple DNA-damaging agents, including temozolomide (TMZ). We investigated ABT-888+TMZ combination therapy in multiple xenograft models representing various human tumors having different responses to TMZ. EXPERIMENTAL DESIGN: ABT-888+TMZ efficacy in xenograft tumors implanted in subcutaneous, orthotopic, and metastatic sites was assessed by tumor burden, expression of poly(ADP-ribose) polymer, and O(6)-methylguanine methyltransferase (MGMT). RESULTS: Varying levels of ABT-888+TMZ sensitivity were evident across a broad histologic spectrum of models (55-100% tumor growth inhibition) in B-cell lymphoma, small cell lung carcinoma, non-small cell lung carcinoma, pancreatic, ovarian, breast, and prostate xenografts, including numerous regressions. Combination efficacy in otherwise TMZ nonresponsive tumors suggests that TMZ resistance may be overcome by poly(ADP-ribose) polymerase inhibition. Profound ABT-888+TMZ efficacy was seen in experimental metastases models that acquired resistance to TMZ. Moreover, TMZ resistance was overcome in crossover treatments, indicating that combination therapy may overcome acquired TMZ resistance. Neither tumor MGMT, mismatch repair, nor poly(ADP-ribose) polymer correlated with the degree of sensitivity to ABT-888+TMZ. CONCLUSIONS: Robust ABT-888+TMZ efficacy is observed across a spectrum of tumor types, including orthotopic and metastatic implantation. As many TMZ nonresponsive tumors proved sensitive to ABT-888+TMZ, this novel combination may broaden the clinical use of TMZ beyond melanoma and glioma. Although TMZ resistance may be influenced by MGMT, neither MGMT nor other mechanisms of TMZ resistance (mismatch repair) precluded sensitivity to ABT-888+TMZ. Underlying mechanisms of TMZ resistance in these models are not completely understood but likely involve mechanisms independent of MGMT.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzimidazóis/administração & dosagem , Dacarbazina/análogos & derivados , Animais , Antineoplásicos Alquilantes/administração & dosagem , Dano ao DNA , Metilases de Modificação do DNA/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Camundongos SCID , Metástase Neoplásica , Transplante de Neoplasias , Temozolomida , Proteínas Supressoras de Tumor/metabolismo
18.
Cancer Biol Ther ; 7(6): 934-41, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18376143

RESUMO

Majority of chemotherapeutic agents inhibit tumor growth by inducing apoptosis or necrosis. The DNA alkylating agent, N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), kills cells by necrosis through massive production of DNA strand breaks and subsequent over-activation of PARP. Inhibition of PARP, either through PARP1 genetic ablation or through small molecule PARP inhibitors, protected MNNG-induced cell death in certain cell types including MEF and primary cortical cultures. We report here that a potent PARP inhibitor, ABT-888, facilitates the induction of apoptotic cell death in HeLa cells treated with MNNG. Although the release of cytochrome c from mitochondria to cytosol was observed in HeLa cells treated with either MNNG alone or the combination of MNNG and ABT-888 (MNNG/ABT-888), apoptosis is observed only in HeLa cells treated with MNNG/ABT-888. Bcl-2 family proteins regulate the release of cytochrome c. Downregulation of Bax and Bak by their corresponding siRNAs or overexpression of Bcl-xl inhibited the release of cytochrome c from mitochondria to cytosol, and inhibited apoptosis induced by MNNG/ABT-888. Further examination indicates that ATP concentration is greatly reduced in HeLa cells treated with MNNG alone, but not in HeLa cells treated with MNNG/ABT-888. Reduction of ATP concentration by F0F1-ATP synthase inhibitor oligomycin A renders HeLa cells resistant to the apoptosis induction by treatment with MNNG/ABT-888. Unlike in HeLa cells, ABT-888 protected MNNG induced cell death in normal human fibroblasts. Our study provides evidence that PARP activity determines the fate of HeLa cells by regulating the level of ATP after treatment with MNNG.


Assuntos
Dano ao DNA , Regulação Enzimológica da Expressão Gênica , Poli(ADP-Ribose) Polimerases/metabolismo , Alquilantes/farmacologia , Animais , Apoptose , Benzimidazóis/farmacologia , Citocromos c/metabolismo , Fibroblastos/metabolismo , Células HeLa , Humanos , Metilnitronitrosoguanidina/farmacologia , Camundongos , NAD/química , Oligomicinas/farmacologia
19.
Bioorg Med Chem ; 15(6): 2441-52, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17258463

RESUMO

Thr-211 is one of three different amino acid residues in the kinase domain of protein kinase B/Akt as compared to protein kinase A (PKA), a closely related analog in the same AGC family. In an attempt to improve the potency and selectivity of our indazole-pyridine series of Akt inhibitors over PKA, efforts have focused on the incorporation of a chemical functionality to interact with the hydroxy group of Thr-211. Several substituents including an oxygen anion, amino, and nitro groups have been introduced at the C-6 position of the indazole scaffold, leading to a significant drop in Akt potency. Incorporation of a nitrogen atom into the phenyl ring at the same position (i.e., 9f) maintained the Akt activity and, in some cases, improved the selectivity over PKA. The structure-activity relationships of the new pyridine-pyrazolopyridine series of Akt inhibitors and their structural features when bound to PKA are also discussed.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Pirazóis/síntese química , Piridinas/síntese química , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Pirazóis/química , Pirazóis/farmacologia , Piridinas/química , Piridinas/farmacologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas/efeitos dos fármacos
20.
Bioorg Med Chem Lett ; 16(6): 1679-85, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16403626

RESUMO

A novel series of Akt/PKB inhibitors derived from a screening lead (1) has been prepared. The novel trans-3,4'-bispyridinylethylenes described herein are potent inhibitors of Akt/PKB with IC(50) values in the low double-digit nanomolar range against Akt1. Compound 2q shows excellent selectivity against distinct families of kinases such as tyrosine kinases and CAMK, and displays poor to modest selectivity against closely related kinases in the AGC and CMGC families. The cellular activities including inhibition of cell growth and phosphorylation of downstream target GSK3 are also described. The X-ray structure of compound 2q complexed with PKA in the ATP binding site was determined.


Assuntos
Antineoplásicos , Inibidores Enzimáticos , Etilenos , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Sítios de Ligação , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Etilenos/síntese química , Etilenos/farmacologia , Quinase 3 da Glicogênio Sintase , Humanos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases/antagonistas & inibidores , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA